1
|
Kong X, Boeckhaus J, Wang F, Shi C, Zhang H, Gross O, Ding J, Zhang Y. A retrospective study of kidney disease in Alport syndrome during and after pregnancy. J Nephrol 2025:10.1007/s40620-025-02252-2. [PMID: 40392259 DOI: 10.1007/s40620-025-02252-2] [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: 06/07/2024] [Accepted: 02/14/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND During pregnancy, hyperfiltration and other factors are hypothesized to contribute to the progression of kidney disease in women with Alport syndrome. To evaluate the status of kidney disease, clinical data from mothers with Alport syndrome in China and Europe over the pregnancy were analyzed. METHODS This retrospective observational study collected data to evaluate proteinuria, kidney function and Alport stage prior to, during, and after pregnancy, respectively. RESULTS A total of 74 women were enrolled, 82% of them with X-linked Alport syndrome and 11% with autosomal Alport syndrome (unknown in 5 patients). Detailed information on the course of pregnancy was available for 62 pregnancies from 52 different women. No fetal malformations were observed. Mean gestational age was 37.9 ± 2.7 weeks (n = 55). Complications included high blood pressure (n = 8), abortion (n = 5), preeclampsia (n = 5), gestational diabetes (n = 3), nephrotic syndrome (n = 2), cervical insufficiency with fetal growth delay (n = 2), premature rupture of membranes (n = 1) and acute intrauterine fetal distress (n = 1). Median proteinuria was 350 (30-2465) mg/day prior to pregnancy, 2390 (450-11,450) mg/day during pregnancy, and 590 (40-2650) mg/day at a mean postpartum follow-up time of 4.5 ± 2.1 years. Mean estimated glomerular filtration rate (eGFR) decreased by 17.2 ± 16.7 ml/min/1.73 m2, from 96.1 ± 32.9 to 78.9 ± 37 ml/min/1.73 m2 after pregnancy (n = 15; p = 0.003). The eGFR loss was higher in women with eGFR < 90 ml/min/1.73 m2 prior to pregnancy compared to women with normal renal function (- 21.5 ± 9.8 vs. - 14 ± 20 ml/min/1.73 m2), and in women with severe variants compared to women with less severe variants (- 21.5 ± 20.2 vs. - 11.3 ± 19.0 ml/min/1.73 m2). Progression of Alport stage after pregnancy was observed in 42% of the women, 31% remained in stage 0-1, and 23% remained in stage 2. CONCLUSIONS This study provides important data on the natural history of Alport syndrome in women who have undergone a pregnancy. Women with severe variants of Alport syndrome, and women with eGFR below 90 ml/min/1.73 m2 face greater risks of kidney disease progression after pregnancy. Further prospective studies are required to confirm these findings.
Collapse
Affiliation(s)
- Xinxin Kong
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi An Men Da Jie, Beijing, 100034, China
| | - Jan Boeckhaus
- Clinic for Nephrology and Rheumatology, University Medicine Goettingen, Robert-Koch Str. 40, 37075, Goettingen, Germany
| | - Fang Wang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi An Men Da Jie, Beijing, 100034, China
| | - Chunyan Shi
- Department of Gynaecology and Obstetrics, Peking University First Hospital, No. 1 Xi An Men Da Jie, Beijing, 100034, China
| | - Hongwen Zhang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi An Men Da Jie, Beijing, 100034, China
| | - Oliver Gross
- Clinic for Nephrology and Rheumatology, University Medicine Goettingen, Robert-Koch Str. 40, 37075, Goettingen, Germany.
| | - Jie Ding
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi An Men Da Jie, Beijing, 100034, China.
| | - Yanqin Zhang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi An Men Da Jie, Beijing, 100034, China.
| |
Collapse
|
2
|
Xie L, Ding Y, Qiu Y, Shi Y. Synergistic toxicity of compound heterozygous mutations in the COL4A3 gene causes end-stage renal disease in A large family of Alport syndrome. Gene 2025; 937:149132. [PMID: 39615805 DOI: 10.1016/j.gene.2024.149132] [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: 06/30/2024] [Revised: 11/17/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
Alport syndrome (AS) is a genetic disorder characterized by kidney disease and hearing/vision abnormalities, resulting from mutations in the COL4A3, COL4A4, or COL4A5 genes. While numerous mutations have been identified in AS cases, the precise molecular mechanisms, particularly for compound mutations, remain under investigation. This study investigated the molecular mechanisms of AS in a proband with end-stage kidney disease (ESKD) using whole exome sequencing, which identified two compound heterozygous COL4A3 missense mutations: NM_000091.5:c.1354G > A (p.G452R) and NM_000091.5:c.4793 T > G (p.L1598R). Sixteen family members of the proband were genotyped, and further analyses, including in silico structural prediction, molecular docking, and in vitro co-immunoprecipitation assays, revealed that the p.G452R mutation disrupted the collagen triple helical structure, associated with hematuria in carriers, while the p.L1598R mutation interfered with the interaction between the NC1 domains of COL4A3 and COL4A4 proteins, crucial for collagen trimerization. These findings demonstrate a synergistic loss-of-function effect of the two mutations, contributing to the AS pathogenesis in the proband, and emphasize the importance of genetic screening and personalized treatment strategies for AS.
Collapse
Affiliation(s)
- Longxin Xie
- Physician-Scientist Program, School of Medicine, Tsinghua University, Beijing 100084, China; Department of Kidney Transplantation, Center of Organ Transplantation, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China.
| | - Yuxi Ding
- Physician-Scientist Program, School of Medicine, Tsinghua University, Beijing 100084, China; Department of Kidney Transplantation, Center of Organ Transplantation, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Ying Qiu
- Physician-Scientist Program, School of Medicine, Tsinghua University, Beijing 100084, China.
| | - Yi Shi
- Department of Kidney Transplantation, Center of Organ Transplantation, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China.
| |
Collapse
|
3
|
Lim C, Lim RS, Choo J, Leow EH, Chan GC, Zhang Y, Ng JL, Chin HL, Tan ES, Goh J, Gandhi N, Ng YH, Than M, Ganesan I, Chong SL, Yap C, Chao SM, Cham B, Kam S, Lim JY, Mok I, Tan HZ, Kwek JL, Lee TL, Wang Z, Goh SM, Lim R, Yeo SC, Teo BW, Da Y, Matchar D, Ng KH. Clinical Implementation of Nephrologist-Led Genomic Testing for Glomerular Diseases in Singapore: Rationale and Protocol. Am J Nephrol 2024; 56:158-171. [PMID: 39626636 PMCID: PMC11975324 DOI: 10.1159/000542942] [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: 10/01/2024] [Accepted: 12/02/2024] [Indexed: 01/14/2025]
Abstract
INTRODUCTION The early diagnosis and appropriate treatment of monogenic glomerular diseases can reduce kidney failure, avoid unnecessary investigations such as kidney biopsies and ineffective treatment with immunosuppressants, guide transplant decisions, and inform the genetic risks of their family members. Yet, genetic testing for kidney disease is underutilized in Singapore. We aimed to implement a nephrologist-led genetic service and evaluate the acceptance, adoption, utility, and cost-effectiveness of genetic testing for monogenic glomerular disease in Singapore. METHODS We will perform a prospective, multi-centre, type II hybrid effectiveness-implementation study with a post-design to evaluate both implementation and clinical outcomes of nephrologist-led genetic testing for suspected genetic glomerular kidney diseases. The multi-disciplinary implementation team will train "genetic nephrologists" to provide pre- and post-test counselling, order targeted exome panel sequencing for suspected glomerular kidney diseases (persistent microscopic haematuria and/or albuminuria or proteinuria in the absence of known causes, steroid-resistant primary nephrotic syndrome, apparent familial IgA nephropathy, or chronic kidney disease with no apparent cause), and interpret genetic test results; create workflows for patient referral, evaluation and management, and discuss genetic results at regular genomic board meetings. The outcomes are acceptance, appropriateness and adoption among patients and nephrologists, utility (proportion of patients who received genetic testing and have a confirmed diagnosis of genetic glomerular disease), and cost-effectiveness. CONCLUSION This study will create and evaluate a nephrologist-led genetic service, develop an efficient variant curation process, and inform future recommendations on the optimal referral and genetic testing strategy for monogenic glomerular disease in Singapore. This will facilitate the future mainstreaming of genetic testing that will enable precision medicine in kidney care.
Collapse
Affiliation(s)
- Cynthia Lim
- Department of Renal Medicine, Medicine, Singapore General Hospital, SingHealth Duke-NUS Academic Medical Center, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ru Sin Lim
- Department of Renal Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - Jason Choo
- Department of Renal Medicine, Medicine, Singapore General Hospital, SingHealth Duke-NUS Academic Medical Center, Singapore, Singapore
| | - Esther Huimin Leow
- Department of Paediatrics, Nephrology, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Gek Cher Chan
- Department of Medicine, Nephrology, National University Hospital, Singapore, Singapore
| | - Yaochun Zhang
- Department of Paediatrics, National University of Singapore, Singapore, Singapore
| | - Jun Li Ng
- Department of Paediatrics, National University of Singapore, Singapore, Singapore
| | - Hui-Lin Chin
- Department of Paediatrics, National University of Singapore, Singapore, Singapore
| | - Ee Shien Tan
- Department of Paediatrics, Genetics, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Jeannette Goh
- Department of Paediatrics, Genetics, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Naline Gandhi
- Program in Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore
| | - Yong Hong Ng
- Department of Paediatrics, Nephrology, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Mya Than
- Department of Paediatrics, National University of Singapore, Singapore, Singapore
| | - Indra Ganesan
- Department of Paediatrics, Nephrology, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Siew Le Chong
- Department of Paediatrics, Nephrology, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Celeste Yap
- Department of Paediatrics, Nephrology, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Sing Ming Chao
- Department of Paediatrics, Nephrology, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Breana Cham
- Department of Paediatrics, Genetics, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Sylvia Kam
- Department of Paediatrics, Genetics, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Jiin Ying Lim
- Department of Paediatrics, Genetics, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Irene Mok
- Department of Renal Medicine, Medicine, Singapore General Hospital, SingHealth Duke-NUS Academic Medical Center, Singapore, Singapore
| | - Hui Zhuan Tan
- Department of Renal Medicine, Medicine, Singapore General Hospital, SingHealth Duke-NUS Academic Medical Center, Singapore, Singapore
| | - Jia Liang Kwek
- Department of Renal Medicine, Medicine, Singapore General Hospital, SingHealth Duke-NUS Academic Medical Center, Singapore, Singapore
| | - Tung Lin Lee
- Department of Renal Medicine, Medicine, Singapore General Hospital, SingHealth Duke-NUS Academic Medical Center, Singapore, Singapore
| | - Ziyin Wang
- Department of Renal Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - Su Mein Goh
- Department of Renal Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - Regina Lim
- Department of Renal Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - See Cheng Yeo
- Department of Renal Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - Boon Wee Teo
- Department of Medicine, Nephrology, National University of Singapore, Singapore, Singapore
| | - Yi Da
- Department of Medicine, Nephrology, National University of Singapore, Singapore, Singapore
| | - David Matchar
- Program in Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore
- Department of Medicine (General Internal Medicine), Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Kar Hui Ng
- Department of Paediatrics, National University of Singapore, Singapore, Singapore
| |
Collapse
|
4
|
Kang E, Park BH, Lee H, Kang HG, Kim JH, Kim YN, Jung Y, Rim H, Shin HS. A comprehensive review of Alport syndrome: definition, pathophysiology, clinical manifestations, and diagnostic considerations. Kidney Res Clin Pract 2024:j.krcp.24.065. [PMID: 39384344 DOI: 10.23876/j.krcp.24.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/22/2024] [Indexed: 10/11/2024] Open
Abstract
Alport syndrome, a rare genetic disorder affecting around 1 in 50,000 individuals, primarily presents as microscopic hematuria and chronic kidney disease (CKD) with associated extrarenal complications. The Alport syndrome results from mutations in COL4A3, COL4A4, and COL4A5 genes, disrupting the formation of the α3-α4-α5 chain in the collagen IV network. The etiology involves X chromosome-related, autosomal dominant, autosomal recessive, and digenic inheritance patterns. The disease primarily manifests as kidney involvement, featuring persistent hematuria, proteinuria, and a progressive decline in renal function. Hearing loss, ocular abnormalities, and extrarenal manifestations further contribute to its complexity. Genotype-phenotype correlations are relatively evident, with distinct presentations in X-linked, autosomal recessive, and autosomal dominant cases. Diagnosis relies on urinalysis, histologic examination, and genetic testing with advancements in next-generation sequencing aiding identification. Although no specific treatment exists, early diagnosis improves outcomes, emphasizing the importance of genetic testing for prognosis and familial screening. The purpose of this review is to advance knowledge and enhance understanding of Alport syndrome.
Collapse
Affiliation(s)
- Eunjeong Kang
- Transplantation Center, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Byung Hwa Park
- Renal Division, Department of Internal Medicine, Kosin University Gospel Hospital, Kosin University College of Medicine, Busan, Republic of Korea
- Transplantation Research Institute, Kosin University College of Medicine, Busan, Republic of Korea
| | - Hajeong Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hee Gyung Kang
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji Hyun Kim
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Ye Na Kim
- Renal Division, Department of Internal Medicine, Kosin University Gospel Hospital, Kosin University College of Medicine, Busan, Republic of Korea
- Transplantation Research Institute, Kosin University College of Medicine, Busan, Republic of Korea
| | - Yeonsoon Jung
- Renal Division, Department of Internal Medicine, Kosin University Gospel Hospital, Kosin University College of Medicine, Busan, Republic of Korea
- Transplantation Research Institute, Kosin University College of Medicine, Busan, Republic of Korea
| | - Hark Rim
- Renal Division, Department of Internal Medicine, Kosin University Gospel Hospital, Kosin University College of Medicine, Busan, Republic of Korea
- Transplantation Research Institute, Kosin University College of Medicine, Busan, Republic of Korea
| | - Ho Sik Shin
- Renal Division, Department of Internal Medicine, Kosin University Gospel Hospital, Kosin University College of Medicine, Busan, Republic of Korea
- Transplantation Research Institute, Kosin University College of Medicine, Busan, Republic of Korea
| |
Collapse
|
5
|
Mou X, Leeman SM, Roye Y, Miller C, Musah S. Fenestrated Endothelial Cells across Organs: Insights into Kidney Function and Disease. Int J Mol Sci 2024; 25:9107. [PMID: 39201792 PMCID: PMC11354928 DOI: 10.3390/ijms25169107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/07/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
In the human body, the vascular system plays an indispensable role in maintaining homeostasis by supplying oxygen and nutrients to cells and organs and facilitating the removal of metabolic waste and toxins. Blood vessels-the key constituents of the vascular system-are composed of a layer of endothelial cells on their luminal surface. In most organs, tightly packed endothelial cells serve as a barrier separating blood and lymph from surrounding tissues. Intriguingly, endothelial cells in some tissues and organs (e.g., choroid plexus, liver sinusoids, small intestines, and kidney glomerulus) form transcellular pores called fenestrations that facilitate molecular and ionic transport across the vasculature and mediate immune responses through leukocyte transmigration. However, the development and unique functions of endothelial cell fenestrations across organs are yet to be fully uncovered. This review article provides an overview of fenestrated endothelial cells in multiple organs. We describe their development and organ-specific roles, with expanded discussions on their contributions to glomerular health and disease. We extend these discussions to highlight the dynamic changes in endothelial cell fenestrations in diabetic nephropathy, focal segmental glomerulosclerosis, Alport syndrome, and preeclampsia, and how these unique cellular features could be targeted for therapeutic development. Finally, we discuss emerging technologies for in vitro modeling of biological systems, and their relevance for advancing the current understanding of endothelial cell fenestrations in health and disease.
Collapse
Affiliation(s)
- Xingrui Mou
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, USA
| | - Sophia M. Leeman
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, USA
- Department of Computer Science, Duke University, Durham, NC 27710, USA
| | - Yasmin Roye
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, USA
| | - Carmen Miller
- Department of Biology, Duke University, Durham, NC 27710, USA
| | - Samira Musah
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27710, USA
- Center for Biomolecular and Tissue Engineering, Duke University, Durham, NC 27710, USA
- Division of Nephrology, Department of Medicine, School of Medicine, Duke University, Durham, NC 27710, USA
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
- Faculty of the Developmental and Stem Cell Biology Program, Duke Regeneration Center, Duke MEDx Initiative, Duke University, Durham, NC 27710, USA
| |
Collapse
|
6
|
Kohan DE, Bedard P, Jenkinson C, Hendry B, Komers R. Mechanism of protective actions of sparsentan in the kidney: lessons from studies in models of chronic kidney disease. Clin Sci (Lond) 2024; 138:645-662. [PMID: 38808486 PMCID: PMC11139641 DOI: 10.1042/cs20240249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/30/2024]
Abstract
Simultaneous inhibition of angiotensin II AT1 and endothelin ETA receptors has emerged as a promising approach for treatment of chronic progressive kidney disease. This therapeutic approach has been advanced by the introduction of sparsentan, the first dual AT1 and ETA receptor antagonist. Sparsentan is a single molecule with high affinity for both receptors. It is US Food and Drug Administration approved for immunoglobulin A nephropathy (IgAN) and is currently being developed as a treatment for rare kidney diseases, such as focal segmental glomerulosclerosis. Clinical studies have demonstrated the efficacy and safety of sparsentan in these conditions. In parallel with clinical development, studies have been conducted to elucidate the mechanisms of action of sparsentan and its position in the context of published evidence characterizing the nephroprotective effects of dual ETA and AT1 receptor inhibition. This review summarizes this evidence, documenting beneficial anti-inflammatory, antifibrotic, and hemodynamic actions of sparsentan in the kidney and protective actions in glomerular endothelial cells, mesangial cells, the tubulointerstitium, and podocytes, thus providing the rationale for the use of sparsentan as therapy for focal segmental glomerulosclerosis and IgAN and suggesting potential benefits in other renal diseases, such as Alport syndrome.
Collapse
Affiliation(s)
- Donald E. Kohan
- Division of Nephrology, University of Utah Health, Salt Lake City, UT, U.S.A
| | | | | | - Bruce Hendry
- Travere Therapeutics, Inc., San Diego, CA, U.S.A
| | - Radko Komers
- Travere Therapeutics, Inc., San Diego, CA, U.S.A
| |
Collapse
|
7
|
Graziani L, Minotti C, Carriero ML, Bengala M, Lai S, Terracciano A, Novelli A, Novelli G. A Novel COL4A5 Pathogenic Variant Joins the Dots in a Family with a Synchronous Diagnosis of Alport Syndrome and Polycystic Kidney Disease. Genes (Basel) 2024; 15:597. [PMID: 38790225 PMCID: PMC11121527 DOI: 10.3390/genes15050597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Alport Syndrome (AS) is the most common genetic glomerular disease, and it is caused by COL4A3, COL4A4, and COL4A5 pathogenic variants. The classic phenotypic spectrum associated with AS ranges from isolated hematuria to chronic kidney disease (CKD) with extrarenal abnormalities. Atypical presentation of the disorder is possible, and it can mislead the diagnosis. Polycystic kidney disease (PKD), which is most frequently associated with Autosomal Dominant PKD (ADPKD) due to PKD1 and PKD2 heterozygous variants, is emerging as a possible clinical manifestation in COL4A3-A5 patients. We describe a COL4A5 novel familial frameshift variant (NM_000495.5: c.1095dup p.(Leu366ValfsTer45)), which was associated with AS and PKD in the hemizygous proband, as well as with PKD, IgA glomerulonephritis and focal segmental glomerulosclerosis (FSGS) in the heterozygous mother. Establishing the diagnosis of AS can sometimes be difficult, especially in the context of misleading family history and atypical phenotypic features. This case study supports the emerging genotypic and phenotypic heterogeneity in COL4A3-A5-associated disorders, as well as the recently described association between PKD and collagen type IV (Col4) defects. We highlight the importance of the accurate phenotyping of all family members and the relevance of next-generation sequencing in the differential diagnosis of hereditary kidney disease.
Collapse
Affiliation(s)
- Ludovico Graziani
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.M.); (M.L.C.); (G.N.)
| | - Chiara Minotti
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.M.); (M.L.C.); (G.N.)
| | - Miriam Lucia Carriero
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.M.); (M.L.C.); (G.N.)
| | - Mario Bengala
- Medical Genetics Unit, Tor Vergata University Hospital, 00133 Rome, Italy;
| | - Silvia Lai
- Division of Nephrology, Department of Translational and Precision Medicine, “Sapienza” University, 00133 Rome, Italy;
| | - Alessandra Terracciano
- Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (A.T.); (A.N.)
| | - Antonio Novelli
- Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (A.T.); (A.N.)
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.M.); (M.L.C.); (G.N.)
- Medical Genetics Unit, Tor Vergata University Hospital, 00133 Rome, Italy;
| |
Collapse
|
8
|
Puapatanakul P, Miner JH. Alport syndrome and Alport kidney diseases - elucidating the disease spectrum. Curr Opin Nephrol Hypertens 2024; 33:283-290. [PMID: 38477333 PMCID: PMC10990029 DOI: 10.1097/mnh.0000000000000983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
PURPOSE OF REVIEW With the latest classification, variants in three collagen IV genes, COL4A3 , COL4A4 , and COL4A5 , represent the most prevalent genetic kidney disease in humans, exhibiting diverse, complex, and inconsistent clinical manifestations. This review breaks down the disease spectrum and genotype-phenotype correlations of kidney diseases linked to genetic variants in these genes and distinguishes "classic" Alport syndrome (AS) from the less severe nonsyndromic genetically related nephropathies that we suggest be called "Alport kidney diseases". RECENT FINDINGS Several research studies have focused on the genotype-phenotype correlation under the latest classification scheme of AS. The historic diagnoses of "benign familial hematuria" and "thin basement membrane nephropathy" linked to heterozygous variants in COL4A3 or COL4A4 are suggested to be obsolete, but instead classified as autosomal AS by recent expert consensus due to a significant risk of disease progression. SUMMARY The concept of Alport kidney disease extends beyond classic AS. Patients carrying pathogenic variants in any one of the COL4A3/A4/A5 genes can have variable phenotypes ranging from completely normal/clinically unrecognizable, hematuria without or with proteinuria, or progression to chronic kidney disease and kidney failure, depending on sex, genotype, and interplays of other genetic as well as environmental factors.
Collapse
Affiliation(s)
- Pongpratch Puapatanakul
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Jeffrey H. Miner
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
9
|
Firat EAM, Buhl EM, Bouteldja N, Smeets B, Eriksson U, Boor P, Klinkhammer BM. PDGF-D Is Dispensable for the Development and Progression of Murine Alport Syndrome. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:641-655. [PMID: 38309427 DOI: 10.1016/j.ajpath.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/18/2023] [Accepted: 12/27/2023] [Indexed: 02/05/2024]
Abstract
Alport syndrome is an inherited kidney disease, which can lead to glomerulosclerosis and fibrosis, as well as end-stage kidney disease in children and adults. Platelet-derived growth factor-D (PDGF-D) mediates glomerulosclerosis and interstitial fibrosis in various models of kidney disease, prompting investigation of its role in a murine model of Alport syndrome. In vitro, PDGF-D induced proliferation and profibrotic activation of conditionally immortalized human parietal epithelial cells. In Col4a3-/- mice, a model of Alport syndrome, PDGF-D mRNA and protein were significantly up-regulated compared with non-diseased wild-type mice. To analyze the therapeutic potential of PDGF-D inhibition, Col4a3-/- mice were treated with a PDGF-D neutralizing antibody. Surprisingly, PDGF-D antibody treatment had no effect on renal function, glomerulosclerosis, fibrosis, or other indices of kidney injury compared with control treatment with unspecific IgG. To characterize the role of PDGF-D in disease development, Col4a3-/- mice with a constitutive genetic deletion of Pdgfd were generated and analyzed. No difference in pathologic features or kidney function was observed in Col4a3-/-Pdgfd-/- mice compared with Col4a3-/-Pdgfd+/+ littermates, confirming the antibody treatment data. Mechanistically, lack of proteolytic PDGF-D activation in Col4a3-/- mice might explain the lack of effects in vivo. In conclusion, despite its established role in kidney fibrosis, PDGF-D, without further activation, does not mediate the development and progression of Alport syndrome in mice.
Collapse
Affiliation(s)
| | - Eva Miriam Buhl
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany; Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany
| | - Nassim Bouteldja
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Bart Smeets
- Department of Pathology, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - Ulf Eriksson
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany; Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany; Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany.
| | | |
Collapse
|
10
|
Deng Z, Zhou Q, Zhou TG. A case report and literature study on Alport syndrome featuring nephrotic syndrome as its primary manifestation. Transpl Immunol 2023; 81:101941. [PMID: 37866673 DOI: 10.1016/j.trim.2023.101941] [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: 07/06/2023] [Revised: 10/07/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
BACKGROUND Historically, due to the lack of distinct clinical symptoms, Alport syndrome, a hereditary kidney disease prevalent in children and a leading cause of kidney failure, has often been misdiagnosed as other kidney conditions. CASE DESCRIPTION This article presents a comprehensive review and analysis of clinical data concerning a child diagnosed with Alport syndrome, where nephrotic syndrome served as the primary manifestation. The male child in this case exhibited symptoms starting at the age of 6, initially diagnosed as nephrotic syndrome. Consequently, oral steroid medication was administered, proving ineffective. Due to persistent proteinuria and microscopic hematuria, a renal biopsy was performed. Immunofluorescence staining revealed no abnormal expression of the α3, α4, and α5 chains of type IV collagen. Notably, electron microscopy revealed the basement membrane to be partially torn and arachnoid. Genetic testing indicated a hemizygous COL4A5 acceptor-splice-site mutation c.4707-1(IVS50)G > A, inherited from his mother. CONCLUSION This specific mutated locus, being the first of its kind reported, adds valuable information to the existing gene mutation spectrum of Alport syndrome. Consequently, it emphasizes the importance for clinicians to deepen their understanding of rare kidney diseases, contributing to enhanced diagnostic accuracy and improved patient care.
Collapse
Affiliation(s)
- Zhuo Deng
- Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Qi Zhou
- Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Tai-Guang Zhou
- Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
11
|
Rhode H, Lüse A, Tautkus B, Nabity M, John-Kroegel U, Weigel F, Dost A, Schitke J, Metzing O, Böckhaus J, Rubel D, Kiess W, Gross O. Urinary Protein-Biomarkers Reliably Indicate Very Early Kidney Damage in Children With Alport Syndrome Independently of Albuminuria and Inflammation. Kidney Int Rep 2023; 8:2778-2793. [PMID: 38106579 PMCID: PMC10719601 DOI: 10.1016/j.ekir.2023.09.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/04/2023] [Accepted: 09/25/2023] [Indexed: 12/19/2023] Open
Abstract
Introduction Alport syndrome (AS) is a hereditary type IV collagen disease. It starts shortly after birth, without clinical symptoms, and progresses to end-stage kidney disease early in life. The earlier therapy starts, the more effectively end-stage kidney disease can be delayed. Clearly then, to ensure preemptive therapy, early diagnosis is an essential prerequisite. Methods To provide early diagnosis, we searched for protein biomarkers (BMs) by mass spectrometry in dogs with AS stage 0. At this very early stage, we identified 74 candidate BMs. Of these, using commercial enzyme-linked immunosorbent assays (ELISAs), we evaluated 27 in dogs and 28 in children, 50 with AS and 104 healthy controls. Results Most BMs from blood appeared as fractions of multiple variants of the same protein, as shown by their chromatographic distribution before mass spectrometry. Blood samples showed only minor differences because ELISAs rarely detect disease-specific variants. However, in urine , several proteins, individually or in combination, were promising indicators of very early and preclinical kidney injury. The BMs with the highest sensitivity and specificity were collagen type XIII, hyaluronan binding protein 2 (HABP2), and complement C4 binding protein (C4BP). Conclusion We generated very strong candidate BMs by our approach of first examining preclinical AS in dogs and then validating these BMs in children at early stages of disease. These BMs might serve for screening purposes for AS before the onset of kidney damage and therefore allow preemptive therapy.
Collapse
Affiliation(s)
- Heidrun Rhode
- Institute of Biochemistry I, Jena University Hospital, Jena, Germany
| | - Alexandra Lüse
- Institute of Biochemistry I, Jena University Hospital, Jena, Germany
| | - Bärbel Tautkus
- Institute of Biochemistry I, Jena University Hospital, Jena, Germany
| | - Mary Nabity
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas, USA
| | | | | | - Axel Dost
- Department of Pediatrics, Jena University Hospital, Jena, Germany
| | - Julia Schitke
- Department of Pediatrics, Jena University Hospital, Jena, Germany
| | - Oliver Metzing
- Department of Pediatrics, Jena University Hospital, Jena, Germany
| | - Jan Böckhaus
- Clinics for Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
| | - Diana Rubel
- Clinics for Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
| | - Wieland Kiess
- Hospital for Children and Adolescents, University of Leipzig, Liebigstr. 20a, 04103 Leipzig, Germany
| | - Oliver Gross
- Clinics for Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
12
|
Zeng M, Di H, Liang J, Liu Z. Effectiveness of renin-angiotensin-aldosterone system blockers in patients with Alport syndrome: a systematic review and meta-analysis. Nephrol Dial Transplant 2023; 38:2485-2493. [PMID: 37218713 DOI: 10.1093/ndt/gfad105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Although renin-angiotensin-aldosterone system (RAAS) blockers have been considered the primary treatment for patients with Alport syndrome (AS) for a decade, there is no comprehensive review with evidence-based analysis evaluating the effectiveness of RAAS blockers in AS. METHODS A systematic review and meta-analysis was performed of published studies that compared outcomes related to disease progression between patients with AS receiving RAAS blockers with those taking non-RAAS treatment. Outcomes were meta-analyzed using the random effects models. Cochrane risk-of-bias, Newcastle-Ottawa Scale and Grading of Recommendations Assessment, Development and Evaluation methodology (GRADE) assessment determined the certainty of evidence. RESULTS A total of eight studies (1182 patients) were included in the analysis. Overall, the risk of bias was low to moderate. Compared with non-RAAS treatment, RAAS blockers could reduce the rate of progression to end-stage kidney disease (ESKD) [four studies; hazard ratio (HR) 0.33, 95% confidence interval (CI) 0.24-0.45; moderate certainty evidence]. After stratified by genetic types, a similar benefit was detected: male X-linked AS (XLAS) (HR 0.32, 95% CI 0.22-0.48), autosomal recessive AS (HR 0.25, 95% CI 0.10-0.62), female XLAS and autosomal dominant AS (HR 0.40, 95% CI 0.21-0.75). In addition, RAAS blockers showed a clear gradient of benefit depending on the stage of disease at the initiation of treatment. CONCLUSION This meta-analysis suggested that RAAS blockers could be considered as a specific therapy to delay of ESKD for AS with any genetic type, especially at the early stage of the disease, and every further more-effective therapy would be advised to be applied on top of this standard of care.
Collapse
Affiliation(s)
- Mengyao Zeng
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Hongling Di
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Ju Liang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
13
|
Adone A, Anjankar A. Alport Syndrome: A Comprehensive Review. Cureus 2023; 15:e47129. [PMID: 38021591 PMCID: PMC10649250 DOI: 10.7759/cureus.47129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Alport syndrome is an genetic disorder that distresses the basement membrane of the kidneys and can also impact other organs, such as the cochlea of the inner ear and eyes. It is characterized by mutation causing abnormalities in the collagen within the basement membrane, which has a crucial role in the filtration process of the kidneys. These abnormalities lead to progressive kidney damage and often result in chronic kidney disease. In some cases of Alport syndrome, the abnormal collagen can also affect the cochlea in the inner ear, leading to sensorineural hearing loss. Additionally, changes in the ocular lens, named anterior lenticonus, can occur, causing vision problems. Alport syndrome can manifest differently among individuals, and its severity can vary. Some people may experience mild symptoms, while others may develop more severe kidney problems, including end-stage renal disease, which may need dialysis or kidney transplant. Treatment for Alport syndrome primarily focuses on managing its symptoms and complications. Regular monitoring of kidney function and blood pressure, along with medications to control hypertension, are crucial aspects of the management plan. In cases of severe kidney damage, kidney transplantation may be necessary. As with any medical condition, early detection and intervention can improve results and quality of life for persons with Alport syndrome. Therefore, if there is a family history of the disorder or any concerning symptoms, it is essential to seek medical attention promptly. Genetic testing can help confirm the diagnosis and identify affected family members, allowing for appropriate monitoring and management.
Collapse
Affiliation(s)
- Avanti Adone
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Ashish Anjankar
- Biochemistry, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
14
|
Pfau K, Gross O, Bemme S, Meyer P, Take P, Boeckhaus J, Holz FG, Feltgen N. [Ocular alterations in patients with Alport syndrome-An update]. DIE OPHTHALMOLOGIE 2023:10.1007/s00347-022-01805-1. [PMID: 36752793 DOI: 10.1007/s00347-022-01805-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/28/2022] [Accepted: 12/21/2022] [Indexed: 02/09/2023]
Abstract
BACKGROUND AND OBJECTIVE Alport syndrome (AS) is a rare hereditary systemic disease that results in alterations of the kidneys, inner ear, and various structures of the eye. It is caused by mutations in one of the genes encoding collagen type IV. In recent years, new and innovative imaging techniques have added characteristics of ocular alterations in AS and provided new insights, including into the pathogenesis of the disease. The aim of this paper is to provide an overview of the current knowledge of ocular changes in AS, as well as to present the Alport ocular pass. METHOD Narrative review article. RESULTS Ocular manifestations of AS include changes in the cornea, lens, and retina. Specifically, posterior polymorphic corneal dystrophy, anterior lenticonus (pathognomonic for AS), and various retinal changes have been described, which have been further characterized in recent years by newer imaging techniques. In particular, foveal changes in AS may present as both a thickened central retina in the context of foveal hypoplasia or a staircase-like thinning of the fovea. Both lesions could provide further insights into the role of type IV collagen in ocular structures. CONCLUSION The AS can manifest in various structures of the eye. The staircase-like changes of the central retina in AS patients indicate the important role of collagen type IV in the homeostasis and regular function of the inner retinal layers. The often mild foveal hypoplasia may provide clues to the role of collagen type IV in retinal embryogenesis. While anterior lenticonus is pathognomonic for AS and can be treated easily by refractive lens exchange, the only option currently available for retinal alterations is close follow-up and, if necessary, treatment of systemic complications of AS.
Collapse
Affiliation(s)
- Kristina Pfau
- Universitäts-Augenklinik Bonn, Bonn, Deutschland.
- Augenklinik, Universitätsspital Basel, Mittlere Str. 91, 4056, Basel, Schweiz.
| | - Oliver Gross
- Klinik für Nephrologie und Rheumatologie, Universitätsmedizin Göttingen, Göttingen, Deutschland
| | | | - Peter Meyer
- Augenklinik, Universitätsspital Basel, Mittlere Str. 91, 4056, Basel, Schweiz
| | - Patricia Take
- Klinik für Nephrologie und Rheumatologie, Universitätsmedizin Göttingen, Göttingen, Deutschland
| | - Jan Boeckhaus
- Klinik für Nephrologie und Rheumatologie, Universitätsmedizin Göttingen, Göttingen, Deutschland
| | - Frank G Holz
- Universitäts-Augenklinik Bonn, Bonn, Deutschland
| | | |
Collapse
|
15
|
Kashtan CE. Genetic testing and glomerular hematuria-A nephrologist's perspective. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:399-403. [PMID: 35775584 PMCID: PMC9796064 DOI: 10.1002/ajmg.c.31987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/25/2022] [Accepted: 06/19/2022] [Indexed: 01/29/2023]
Abstract
Alport syndrome is an inherited disorder of the kidneys that results from variants in three collagen IV genes-COL4A3, COL4A4, and COL4A5. Early diagnosis and pharmacologic intervention can delay the progression of chronic kidney disease and the onset of kidney failure in patients with Alport syndrome. This article describes the evolution of approaches to the diagnosis and early treatment of Alport syndrome.
Collapse
Affiliation(s)
- Clifford E. Kashtan
- Division of Pediatric NephrologyUniversity of Minnesota Medical SchoolMinneapolisMinnesotaUSA
| |
Collapse
|
16
|
The causes and consequences of paediatric kidney disease on adult nephrology care. Pediatr Nephrol 2022; 37:1245-1261. [PMID: 34389906 DOI: 10.1007/s00467-021-05182-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 05/29/2021] [Accepted: 06/01/2021] [Indexed: 10/20/2022]
Abstract
Adult nephrologists often look after patients who have been diagnosed with kidney disease in childhood. This does present unique challenges to the adult nephrologist, who may be unfamiliar with the underlying cause of kidney disease as well as the complications of chronic kidney disease (CKD) that may have accumulated during childhood. This review discusses common causes of childhood CKD, in particular congenital anomalies of the kidney and urinary tract (CAKUT), autosomal dominant tubulointerstitial kidney disease (ADTKD), polycystic kidney disease, hereditary stone disease, nephrotic syndrome and atypical haemolytic uraemic syndrome. The long-term consequences of childhood CKD, such as the cardiovascular consequences, cognition and education as well as bone health, nutrition and growth are also discussed.
Collapse
|
17
|
Kashtan CE. What the Adult Nephrologist Should Know About Alport Syndrome. Adv Chronic Kidney Dis 2022; 29:225-230. [PMID: 36084969 DOI: 10.1053/j.ackd.2021.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 11/11/2022]
Abstract
Recent trends in the diagnosis, treatment, and classification of collagen IV-associated kidney disease are likely to result in increasing numbers of people in adult nephrology practices who have a confirmed diagnosis of Alport syndrome. These trends include the increasing use of genetic testing in the diagnostic evaluation of people with hematuria, focal segmental glomerulosclerosis, and chronic kidney disease of unknown etiology; early treatment with inhibitors of the renin-angiotensin-aldosterone system to delay kidney failure; and application of an expanded definition of Alport syndrome based on genotype rather than phenotype. This commentary discusses these trends and their implications for the adult nephrologist.
Collapse
Affiliation(s)
- Clifford E Kashtan
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN.
| |
Collapse
|
18
|
Daga S, Ding J, Deltas C, Savige J, Lipska-Ziętkiewicz BS, Hoefele J, Flinter F, Gale DP, Aksenova M, Kai H, Perin L, Barua M, Torra R, Miner JH, Massella L, Ljubanović DG, Lennon R, Weinstock AB, Knebelmann B, Cerkauskaite A, Gear S, Gross O, Turner AN, Baldassarri M, Pinto AM, Renieri A. The 2019 and 2021 International Workshops on Alport Syndrome. Eur J Hum Genet 2022; 30:507-516. [PMID: 35260866 PMCID: PMC8904161 DOI: 10.1038/s41431-022-01075-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 02/04/2022] [Accepted: 02/14/2022] [Indexed: 01/27/2023] Open
Affiliation(s)
- Sergio Daga
- Medical Genetics, University of Siena, Siena, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Jie Ding
- Peking University First Hospital, Beijing, China
| | - Constantinos Deltas
- Biobank.cy Center of Excellence in Biobanking and Biomedical Research and University of Cyprus Medical School, Nicosia, Cyprus
| | - Judy Savige
- Department of Medicine, Melbourne and Northern Health, The University of Melbourne, Parkville, VIC, 3050, Australia
| | - Beata S Lipska-Ziętkiewicz
- Rare Diseases Centre, Clinical Genetics Unit, Department of Biology and Medical Genetics, Medical University of Gdańsk, Gdansk, Poland
| | - Julia Hoefele
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Frances Flinter
- Department of Clinical Genetics, Guys' and St Thomas' NHS Foundation Trust, London, UK
| | - Daniel P Gale
- Department of Renal Medicine, University College London, London, UK
- Rare Renal Disease Registry, UK Renal Registry, Bristol, UK
| | - Marina Aksenova
- Y. Veltischev Research and Clinical Institute for Pediatrics at the Pirogov Russian National Research Medical University, Taldomskaya Street, 2, Moscow, 125412, Russia
| | - Hirofumi Kai
- Department of Molecular Medicine, Kumamoto University, Kumamoto, Japan
| | - Laura Perin
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Moumita Barua
- Toronto General Hospital, Toronto General Research Institute, University of Toronto, Toronto, ON, Canada
| | - Roser Torra
- Inherited Kidney Diseases, Nephrology Department, Fundació Puigvert, IIB-Sant Pau, Medicine Department, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Jeff H Miner
- Division of Nephrology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Laura Massella
- Division of Nephrology, Department of Pediatric Subspecialties, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Danica Galešić Ljubanović
- University of Zagreb School of Medicine, Department of Pathology and Department of Nephropathology and Electron Microscopy Dubrava University Hospital, Zagreb, Croatia
| | - 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 Academic Health Science Centre, Manchester, UK
| | | | - Bertrand Knebelmann
- Nephrology Department, Reference Center for Inherited Kidney Diseases (MARHEA), APHP, Necker Hospital, Paris University, Paris, France
| | - Agne Cerkauskaite
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | | | - Oliver Gross
- Department of Nephrology and Rheumatology, University Medicine Goettingen, Gottingen, Germany
| | - A Neil Turner
- Centre for Inflammation, University of Edinburgh, Edinburgh, UK
| | - Margherita Baldassarri
- Medical Genetics, University of Siena, Siena, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Anna Maria Pinto
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Alessandra Renieri
- Medical Genetics, University of Siena, Siena, Italy.
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy.
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy.
| |
Collapse
|
19
|
Rubel D, Boulanger J, Craciun F, Xu EY, Zhang Y, Phillips L, Callahan M, Weber W, Song W, Ngai N, Bukanov NO, Shi X, Hariri A, Husson H, Ibraghimov-Beskrovnaya O, Liu S, Gross O. Anti-microRNA-21 Therapy on Top of ACE Inhibition Delays Renal Failure in Alport Syndrome Mouse Models. Cells 2022; 11:cells11040594. [PMID: 35203245 PMCID: PMC8869926 DOI: 10.3390/cells11040594] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/28/2022] [Accepted: 02/03/2022] [Indexed: 02/01/2023] Open
Abstract
Col4a3−/− Alport mice serve as an animal model for renal fibrosis. MicroRNA-21 (miR-21) expression has been shown to be increased in the kidneys of Alport syndrome patients. Here, we investigated the nephroprotective effects of Lademirsen anti-miR-21 therapy. We used a fast-progressing Col4a3−/− mouse model with a 129/SvJ background and an intermediate-progressing F1 hybrid mouse model with a mixed genetic background, with angiotensin-converting enzyme inhibitor (ACEi) monotherapy in combination with anti-miR-21 therapy. In the fast-progressing model, the anti miR-21 and ACEi therapies showed an additive effect in the reduction in fibrosis, the decline of proteinuria, the preservation of kidney function and increased survival. In the intermediate-progressing F1 model, the anti-miR-21 and ACEi therapies individually improved kidney pathology. Both also improved kidney function and survival; however, the combination showed a significant additive effect, particularly for survival. RNA sequencing (RNA-seq) gene expression profiling revealed that the anti-miR-21 and ACEi therapies modulate several common pathways. However, anti-miR-21 was particularly effective at normalizing the expression profiles of the genes involved in renal tubulointerstitial injury pathways. In conclusion, significant additive effects were detected for the combination of anti-miR-21 and ACEi therapies on kidney function, pathology and survival in Alport mouse models, as well as a strong differential effect of anti-miR-21 on the renal expression of fibrotic factors. These results support the addition of anti-miR-21 to the current standard of care (ACEi) in ongoing clinical trials in patients with Alport syndrome.
Collapse
Affiliation(s)
- Diana Rubel
- Clinic for Nephrology and Rheumatology, University Medical Center Goettingen, 37075 Goettingen, Germany; (D.R.); (Y.Z.)
| | | | - Florin Craciun
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
| | - Ethan Y. Xu
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
- Excision BioTherapeutics, San Francisco, CA 94111, USA
| | - Yanqin Zhang
- Clinic for Nephrology and Rheumatology, University Medical Center Goettingen, 37075 Goettingen, Germany; (D.R.); (Y.Z.)
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - Lucy Phillips
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
- Abbvie Bioresearch Center, Worcester, MA 01605, USA
| | - Michelle Callahan
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
| | - William Weber
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
- Takeda Pharmaceuticals, Cambridge, MA 02139, USA
| | - Wenping Song
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
| | - Nicholas Ngai
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
| | - Nikolay O. Bukanov
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
- Janssen Pharmaceuticals, Boston, MA 02115, USA
| | - Xingyi Shi
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
- Novartis Institute for BioMedical Research, Boston, MA 02139, USA
| | - Ali Hariri
- Sanofi-Genzyme, Clinical Development, Cambridge, MA 02142, USA; (A.H.); (S.L.)
- Eloxx Pharmaceuticals, Watertown, MA 02140, USA
| | - Hervé Husson
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
| | - Oxana Ibraghimov-Beskrovnaya
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
- Dyne Therapeutics, Waltham, MA 02451, USA
| | - Shiguang Liu
- Sanofi-Genzyme, Clinical Development, Cambridge, MA 02142, USA; (A.H.); (S.L.)
| | - Oliver Gross
- Clinic for Nephrology and Rheumatology, University Medical Center Goettingen, 37075 Goettingen, Germany; (D.R.); (Y.Z.)
- Correspondence: ; Tel.: +49-551-39-60488
| |
Collapse
|
20
|
Oduware E, Iduoriyekemwen NJ, Ibadin M, Aikhionbare H. A Case Report of COL4A5 Gene Mutation Alport Syndrome in 2 Native African Children. Case Rep Nephrol Dial 2021; 11:308-313. [PMID: 34901198 PMCID: PMC8613556 DOI: 10.1159/000519076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/09/2021] [Indexed: 11/19/2022] Open
Abstract
Alport syndrome is a heterogeneous genetic disease involving the basement membrane of the glomeruli, inner ear, retina, and lens capsule. It typically manifests as progressive glomerulopathy that frequently results in end-stage renal disease, high-tone sensorineural deafness, and ocular abnormalities of anterior lenticonus and yellow and white dots and flecks on the macular of the retina. In this report, we describe the cases of 2 siblings: 15- and 13-year-old boys of pure African descent with the COL4A5 gene mutation. Both children had the classical features of Alport syndrome haematuria, proteinuria, progressive sensorineural high-tone hearing loss, and ocular abnormalities. Their renal abnormalities initially regressed on therapy with angiotensin-converting enzyme inhibitors but reoccurred, depicting the need for early diagnosis as the early institution of this therapy before significant glomerulopathy is advocated.
Collapse
Affiliation(s)
- Emmanuel Oduware
- Department of Family Medicine, University of Benin Teaching Hospital, Benin City, Nigeria
| | | | - Michael Ibadin
- Department of Child Health, University of Benin/University of Benin Teaching Hospital, Benin City, Nigeria
| | - Henry Aikhionbare
- Department of Child Health, University of Benin/University of Benin Teaching Hospital, Benin City, Nigeria
| |
Collapse
|
21
|
Smetana J, Vallova V, Wayhelova M, Hladilkova E, Filkova H, Horinova V, Broz P, Mikulasova A, Gaillyova R, Kuglík P. Case Report: Contiguous Xq22.3 Deletion Associated with ATS-ID Syndrome: From Genotype to Further Delineation of the Phenotype. Front Genet 2021; 12:750110. [PMID: 34777475 PMCID: PMC8585740 DOI: 10.3389/fgene.2021.750110] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
Alport syndrome with intellectual disability (ATS-ID, AMME complex; OMIM #300194) is an X-linked contiguous gene deletion syndrome associated with an Xq22.3 locus mainly characterized by hematuria, renal failure, hearing loss/deafness, neurodevelopmental disorder (NDD), midface retrusion, and elliptocytosis. It is thought that ATS-ID is caused by the loss of function of COL4A5 (ATS) and FACL4 (ACSL4) genes through the interstitial (micro)deletion of chromosomal band Xq22.3. We report detailed phenotypic description and results from genome-wide screening of a Czech family with diagnosis ATS-ID (proband, maternal uncle, and two female carriers). Female carriers showed mild clinical features of microscopic hematuria only, while affected males displayed several novel clinical features associated with ATS-ID. Utilization of whole-exome sequencing discovered the presence of approximately 3 Mb of deletion in the Xq23 area, which affected 19 genes from TSC22D3 to CHRDL1. We compared the clinical phenotype with previously reported three ATS-ID families worldwide and correlated their clinical manifestations with the incidence of genes in both telomeric and centromeric regions of the deleted chromosomal area. In addition to previously described phenotypes associated with aberrations in AMMECR1 and FACL4, we identified two genes, members of tripartite motif family MID2 and subunit of the proteasome PA700/19S complex (PSMD10), respectively, as prime candidate genes responsible for additional clinical features observed in our patients with ATS-ID. Overall, our findings further improve the knowledge about the clinical impact of Xq23 deletions and bring novel information about phenotype/genotype association of this chromosomal aberration.
Collapse
Affiliation(s)
- Jan Smetana
- Department of Genetics and Molecular Biology, Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech
| | - Vladimira Vallova
- Department of Genetics and Molecular Biology, Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech.,Department of Medical Genetics and Genomics, University Hospital Brno, Brno, Czech
| | - Marketa Wayhelova
- Department of Genetics and Molecular Biology, Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech.,Department of Medical Genetics and Genomics, University Hospital Brno, Brno, Czech
| | - Eva Hladilkova
- Department of Medical Genetics and Genomics, University Hospital Brno, Brno, Czech
| | - Hana Filkova
- Department of Medical Genetics and Genomics, University Hospital Brno, Brno, Czech
| | | | - Petr Broz
- Department of Biology and Medical Genetics, 2nd Faculty of Medicine, Charles University Prague and Faculty Hospital Motol, Prague, Czech
| | - Aneta Mikulasova
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Renata Gaillyova
- Department of Medical Genetics and Genomics, University Hospital Brno, Brno, Czech
| | - Petr Kuglík
- Department of Genetics and Molecular Biology, Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech.,Department of Medical Genetics and Genomics, University Hospital Brno, Brno, Czech
| |
Collapse
|
22
|
McEwen ST, Rheault MN. Glomerular disease in children: when to biopsy. Nephrol Dial Transplant 2021; 36:1803-1805. [PMID: 31951262 DOI: 10.1093/ndt/gfz280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Indexed: 12/29/2022] Open
Affiliation(s)
- Scott T McEwen
- Department of Pediatrics, Division of Pediatric Nephrology, University of Minnesota Masonic Children's Hospital, Minneapolis, MN, USA
| | - Michelle N Rheault
- Department of Pediatrics, Division of Pediatric Nephrology, University of Minnesota Masonic Children's Hospital, Minneapolis, MN, USA
| |
Collapse
|
23
|
Savige J, Storey H, Watson E, Hertz JM, Deltas C, Renieri A, 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, Lipska-Ziętkiewicz BS. Consensus statement on standards and guidelines for the molecular diagnostics of Alport syndrome: refining the ACMG criteria. Eur J Hum Genet 2021; 29:1186-1197. [PMID: 33854215 PMCID: PMC8384871 DOI: 10.1038/s41431-021-00858-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 02/13/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
The recent Chandos House meeting of the Alport Variant Collaborative extended the indications for screening for pathogenic variants in the COL4A5, COL4A3 and COL4A4 genes beyond the classical Alport phenotype (haematuria, renal failure; family history of haematuria or renal failure) to include persistent proteinuria, steroid-resistant nephrotic syndrome, focal and segmental glomerulosclerosis (FSGS), familial IgA glomerulonephritis and end-stage kidney failure without an obvious cause. The meeting refined the ACMG criteria for variant assessment for the Alport genes (COL4A3-5). It identified 'mutational hotspots' (PM1) in the collagen IV α5, α3 and α4 chains including position 1 Glycine residues in the Gly-X-Y repeats in the intermediate collagenous domains; and Cysteine residues in the carboxy non-collagenous domain (PP3). It considered that 'well-established' functional assays (PS3, BS3) were still mainly research tools but sequencing and minigene assays were commonly used to confirm splicing variants. It was not possible to define the Minor Allele Frequency (MAF) threshold above which variants were considered Benign (BA1, BS1), because of the different modes of inheritances of Alport syndrome, and the occurrence of hypomorphic variants (often Glycine adjacent to a non-collagenous interruption) and local founder effects. Heterozygous COL4A3 and COL4A4 variants were common 'incidental' findings also present in normal reference databases. The recognition and interpretation of hypomorphic variants in the COL4A3-COL4A5 genes remains a challenge.
Collapse
Affiliation(s)
- Judy Savige
- grid.1008.90000 0001 2179 088XDepartment of Medicine (MH and NH), The University of Melbourne, Parkville, VIC Australia
| | - Helen Storey
- grid.239826.40000 0004 0391 895XMolecular Genetics, Viapath Laboratories, Guy’s Hospital, London, UK
| | - Elizabeth Watson
- Elizabeth Watson, South West Genomic Laboratory Hub, North Bristol Trust, Bristol, UK
| | - Jens Michael Hertz
- grid.7143.10000 0004 0512 5013Jens Michael Hertz, Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Constantinos Deltas
- grid.6603.30000000121167908Center of Excellence in Biobanking and Biomedical Research and Molecule Medicine Center, University of Cyprus, Nicosia, Cyprus
| | - Alessandra Renieri
- grid.9024.f0000 0004 1757 4641Medical Genetics, University of Siena, Siena, Italy
| | - Francesca Mari
- Institute de Pathologie et de Genetique ASBL, Departement de Biologie Moleculaire, Gosselies, Belgium
| | - Pascale Hilbert
- Institute de Pathologie et de Genetique ASBL, Departement de Biologie Moleculaire, Gosselies, Belgium
| | - Pavlina Plevova
- grid.412727.50000 0004 0609 0692Department of Medical Genetics, and Department of Biomedical Sciences, University Hospital of Ostrava, Ostrava, Czech Republic
| | - Peter Byers
- grid.34477.330000000122986657Departments of Pathology and Medicine (Medical Genetics), University of Washington, Seattle, WA USA
| | - Agne Cerkauskaite
- grid.6441.70000 0001 2243 2806Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Martin Gregory
- grid.223827.e0000 0001 2193 0096Division of Nephrology, Department of Medicine, University of Utah Health, Salt Lake City, UT USA
| | - Rimante Cerkauskiene
- grid.6441.70000 0001 2243 2806Clinic of Pediatrics, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Danica Galesic Ljubanovic
- grid.412095.b0000 0004 0631 385XDepartment of Pathology, University of Zagreb, School of Medicine, Dubrava University Hospital, Zagreb, Croatia
| | - Francesca Becherucci
- grid.411477.00000 0004 1759 0844Nephrology Unit and Meyer Children’s University Hospital, Firenze, Italy
| | - Carmela Errichiello
- grid.411477.00000 0004 1759 0844Nephrology Unit and Meyer Children’s University Hospital, Firenze, Italy
| | - Laura Massella
- grid.414125.70000 0001 0727 6809Division of Nephrology and Dialysis, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Valeria Aiello
- grid.6292.f0000 0004 1757 1758Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, S. Orsola Hospital, University of Bologna, Bologna, Italy
| | - Rachel Lennon
- grid.5379.80000000121662407Wellcome 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, UK
| | - Louise Hopkinson
- grid.5379.80000000121662407Wellcome 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, UK
| | - Ania Koziell
- grid.13097.3c0000 0001 2322 6764School of Immunology and Microbial Sciences, Faculty of Life Sciences, King’s College London, London, UK
| | - Adrian Lungu
- grid.415180.90000 0004 0540 9980Fundeni Clinical Institute, Pediatric Nephrology Department, Bucharest, Romania
| | | | - Julia Hoefele
- grid.6936.a0000000123222966Institute of Human Genetics, Technical University of Munich, München, Germany
| | | | - Tamara Nikuseva Martic
- grid.4808.40000 0001 0657 4636Department of Biology, School of Medicine University of Zagreb, Zagreb, Croatia
| | - Asheeta Gupta
- grid.415246.00000 0004 0399 7272Birmingham Children’s Hospital, Birmingham, UK
| | - Albertien van Eerde
- grid.5477.10000000120346234Departments of Genetics and Center for Molecular Medicine, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | | | - Samuela Landini
- grid.8404.80000 0004 1757 2304Medical Genetics Unit, Department of Clinical and Experimental Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Viviana Palazzo
- grid.411477.00000 0004 1759 0844Medical Genetics Unit, Meyer Children’s University Hospital, Florence, Italy
| | - Laith al-Rabadi
- grid.223827.e0000 0001 2193 0096Health Sciences Centre, University of UTAH, Salt Lake City, UT USA
| | - Kathleen Claes
- grid.410569.f0000 0004 0626 3338Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Anniek Corveleyn
- grid.410569.f0000 0004 0626 3338Center for Human Genetics, University Hospitals and KU Leuven, Leuven, Belgium
| | - Evelien Van Hoof
- grid.410569.f0000 0004 0626 3338Center for Human Genetics, University Hospitals and KU Leuven, Leuven, Belgium
| | - Micheel van Geel
- grid.412966.e0000 0004 0480 1382Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Maggie Williams
- grid.416201.00000 0004 0417 1173Bristol Genetics Laboratory Pathology Sciences, Southmead Hospital, Bristol, UK
| | - Emma Ashton
- grid.420468.cNorth East Thames Regional Genetics Laboratory, Great Ormond Street Hospital, London, UK
| | - Hendica Belge
- grid.10417.330000 0004 0444 9382Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Elisabeth Ars
- grid.7080.f0000 0001 2296 0625Inherited Kidney Disorders, Fundacio Puigvert, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Agnieszka Bierzynska
- grid.5337.20000 0004 1936 7603Bristol Renal Unit, Bristol Medical School, University of Bristol, Bristol, UK
| | - Concetta Gangemi
- grid.411475.20000 0004 1756 948XDivision of Nephrology and Dialysis, University Hospital of Verona, Verona, Italy
| | - Beata S. Lipska-Ziętkiewicz
- grid.11451.300000 0001 0531 3426Centre for Rare Diseases, and Clinical Genetics Unit, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
24
|
Lusco MA, Fogo AB. Hereditary Nephritis and Thin Glomerular Basement Membrane Lesion. GLOMERULAR DISEASES 2021; 1:135-144. [PMID: 36751492 PMCID: PMC9677744 DOI: 10.1159/000516744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/21/2021] [Indexed: 11/19/2022]
Abstract
Hereditary nephritis (HN) and thin glomerular basement membrane (GBM) lesion share a common clinical presentation of persistent hematuria, thin GBM by kidney biopsy electron microscopic examination, and a mutation in type IV collagen. However, the clinical course and treatment for these entities are different with varying patterns of heredity. Ultrastructural examination of a renal biopsy specimen is essential for the morphologic diagnosis of HN and thin GBM lesion, whereas light microscopy may only give limited diagnostic clues. Additional workup including immunostaining for subtypes of type IV collagen may provide further information on underlying genetic mutations. The diagnosis of HN may lead to treatment with renin-angiotensin system blockade in patients at risk of early-onset renal failure to delay progression to end-stage renal disease. Additionally, patients with isolated microscopic hematuria and thin GBM lesion are at increased risk for chronic kidney disease when associated with other comorbidities; those patients should receive regular clinical assessment to prevent renal function decline.
Collapse
|
25
|
Kortum AJ, Bazelle J, Gomez Selgas A, Kent ACC, Williams TL, Herrtage ME. A retrospective study of canine idiopathic renal haematuria: clinical findings and outcome following medical treatment. J Small Anim Pract 2021; 62:850-860. [PMID: 34075582 DOI: 10.1111/jsap.13352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/16/2021] [Accepted: 04/18/2021] [Indexed: 01/13/2023]
Abstract
OBJECTIVES To characterise and document the progression of idiopathic renal haematuria in a large cohort of medically managed UK dogs. MATERIALS AND METHODS Retrospective study of 41 client-owned dogs with confirmed (n=14), or suspected (n=27) idiopathic renal haematuria from 4 UK-based referral centres. Clinical findings and outcomes of dogs (2001 to 2018) were determined from the review of medical records and telephone follow-up. RESULTS Median survival time from diagnosis was long [1482 (152 to 1825) days] irrespective of treatment and clinical response. Only 1 case was euthanased due to idiopathic renal haematuria, and anaemia or azotaemia occurred infrequently. In total, 25 dogs received angiotensin-converting enzyme-inhibitor or angiotensin receptor blocker therapy, of which 23 received benazepril [0.44 (0.19 to 0.82) mg/kg/24 hours], two received enalapril (0.40 and 0.78 mg/kg/24 hours) and one received telmisartan (1 mg/kg/24 hours). In cases with follow-up urinalyses, complete resolution of haematuria was documented in eight of 19 (42%) dogs following angiotensin-converting enzyme-inhibitor/angiotensin receptor blocker treatment, with partial improvement in five of 19 (26%) and no improvement in six of 19 (31%). Conversely, of the two untreated dogs where outcome was available, one had partial improvement and the other had no improvement. CLINICAL SIGNIFICANCE In this study, idiopathic renal haematuria was associated with a good prognosis and low complication rate. Resolution or improvement in haematuria occurred in both angiotensin-converting enzyme-inhibitor/angiotensin receptor blocker-treated and untreated dogs, indicating that further studies are required to evaluate the effectiveness and safety of these interventions.
Collapse
Affiliation(s)
- A J Kortum
- The Queen's Veterinary School Hospital, Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - J Bazelle
- Davies Veterinary Specialists, Manor Farm Business Park, Higham Gobion, Hitchin, SG5 3HR, UK
| | - A Gomez Selgas
- Animal Health Trust, Lanwades Park, Kentford, Suffolk, CB8 7UU, UK
| | - A C C Kent
- Willows Veterinary Centre and Referral Service, Highlands Road, Shirley, Solihull, West Midlands, B90 4NH, UK
| | - T L Williams
- The Queen's Veterinary School Hospital, Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - M E Herrtage
- The Queen's Veterinary School Hospital, Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| |
Collapse
|
26
|
Takemon Y, Wright V, Davenport B, Gatti DM, Sheehan SM, Letson K, Savage HS, Lennon R, Korstanje R. Uncovering Modifier Genes of X-Linked Alport Syndrome Using a Novel Multiparent Mouse Model. J Am Soc Nephrol 2021; 32:1961-1973. [PMID: 34045313 PMCID: PMC8455275 DOI: 10.1681/asn.2020060777] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 03/27/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Mutations in COL4A5 are responsible for 80% of cases of X-linked Alport Syndrome (XLAS). Although genes that cause AS are well characterized, people with AS who have similar genetic mutations present with a wide variation in the extent of kidney impairment and age of onset, suggesting the activities of modifier genes. METHODS We created a cohort of genetically diverse XLAS male and female mice using the Diversity Outbred mouse resource and measured albuminuria, GFR, and gene expression. Using a quantitative trait locus approach, we mapped modifier genes that can best explain the underlying phenotypic variation measured in our diverse population. RESULTS Genetic analysis identified several loci associated with the variation in albuminuria and GFR, including a locus on the X chromosome associated with X inactivation and a locus on chromosome 2 containing Fmn1. Subsequent analysis of genetically reduced Fmn1 expression in Col4a5 knockout mice showed a decrease in albuminuria, podocyte effacement, and podocyte protrusions in the glomerular basement membrane, which support the candidacy of Fmn1 as a modifier gene for AS. CONCLUSION With this novel approach, we emulated the variability in the severity of kidney phenotypes found in human patients with Alport Syndrome through albuminuria and GFR measurements. This approach can identify modifier genes in kidney disease that can be used as novel therapeutic targets.
Collapse
Affiliation(s)
| | | | - Bernard Davenport
- 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 Academic Health Science Centre, Manchester, United Kingdom
| | | | | | | | | | - 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 Academic Health Science Centre, Manchester, United Kingdom
| | | |
Collapse
|
27
|
Kashtan C. Multidisciplinary Management of Alport Syndrome: Current Perspectives. J Multidiscip Healthc 2021; 14:1169-1180. [PMID: 34045864 PMCID: PMC8149282 DOI: 10.2147/jmdh.s284784] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 04/30/2021] [Indexed: 12/11/2022] Open
Abstract
Alport syndrome is a multisystem disorder that universally affects the kidney and frequently involves the inner ear and the eye. Over the course of a lifetime, addressing the health care needs of a person with Alport syndrome and their family entails the services of primary providers, nephrologists, genetic counselors, audiologists, ophthalmologists, transplant physicians, kidney dieticians, and social workers as well as other healthcare professionals. This article attempts to provide context and guidance regarding the multidisciplinary care of Alport syndrome based on the natural history of the condition.
Collapse
Affiliation(s)
- Clifford Kashtan
- Department of Pediatrics, Division of Pediatric Nephrology, University of Minnesota Medical School, Minneapolis, MN, 55454, USA
| |
Collapse
|
28
|
Long-term outcome among females with Alport syndrome from a single pediatric center. Pediatr Nephrol 2021; 36:945-951. [PMID: 33048202 PMCID: PMC7914153 DOI: 10.1007/s00467-020-04748-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/23/2020] [Accepted: 08/27/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Alport syndrome (AS) is a multisystem condition which can result in progressive kidney disease, hearing loss, and ocular changes. X-linked inheritance is observed in 85% of affected individuals. As a result, most prior studies have focused on males. Girls with AS can also be symptomatic although historically thought to have few clinical manifestations in childhood. The objective of the study was to describe the clinical presentation and course of females with AS. METHODS A single-center retrospective study of all young females with AS between January 1, 1987, and May 20, 2019. Subjects were identified using ICD-9/10 diagnosis codes for AS, familial hematuria, or nephritis. Clinical data were extracted by retrospective chart review. RESULTS Thirty-six female patients were included in the analysis. Mean age at presentation was 5.58 ± 3.0 years, and mean follow-up was 5.9 ± 3.9 years. Twenty-nine patients (80%) had a family history of AS. At end of the follow-up period, gross hematuria was observed in 15 patients (42%), 20 (56%) developed proteinuria, and 2 (6.7%) had an estimated glomerular filtration rate (eGFR) < 90 ml/min/1.73m2 with one patient developing stage 5 chronic kidney disease. Four of the twenty-seven (14.8%) who underwent audiologic testing had an abnormal exam. CONCLUSIONS Known family histories of AS or gross hematuria were the most common reasons for the initial presentation in our cohort. Development of proteinuria, eGFR < 90 ml/min/1.73m2, and abnormal audiology exam are not exceptional findings, suggesting that close monitoring of young females into adulthood is warranted.
Collapse
|
29
|
Chertow GM, Appel GB, Andreoli S, Bangalore S, Block GA, Chapman AB, Chin MP, Gibson KL, Goldsberry A, Iijima K, Inker LA, Knebelmann B, Mariani LH, Meyer CJ, Nozu K, O'Grady M, Silva AL, Stenvinkel P, Torra R, Warady BA, Pergola PE. Study Design and Baseline Characteristics of the CARDINAL Trial: A Phase 3 Study of Bardoxolone Methyl in Patients with Alport Syndrome. Am J Nephrol 2021; 52:180-189. [PMID: 33789284 DOI: 10.1159/000513777] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/03/2020] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Alport syndrome is a rare genetic disorder that affects as many as 60,000 persons in the USA and a total of 103,000 persons (<5 per 10,000) in the European Union [1, 2]. It is the second most common inherited cause of kidney failure and is characterized by progressive loss of kidney function that often leads to end-stage kidney disease. Currently, there are no approved disease-specific agents for therapeutic use. We designed a phase 3 study (CARDINAL; NCT03019185) to evaluate the safety, tolerability, and efficacy of bardoxolone methyl in patients with Alport syndrome. METHODS The CARDINAL phase 3 study is an international, multicenter, double-blind, placebo-controlled, randomized registrational trial. Eligible patients were of ages 12-70 years with confirmed genetic or histologic diagnosis of Alport syndrome, eGFR 30-90 mL/min/1.73 m2, and urinary albumin to creatinine ratio (UACR) ≤3,500 mg/g. Patients with B-type natriuretic peptide values >200 pg/mL at baseline or with significant cardiovascular histories were excluded. Patients were randomized 1:1 to bardoxolone methyl or placebo, with stratification by baseline UACR. RESULTS A total of 371 patients were screened, and 157 patients were randomly assigned to receive bardoxolone methyl (n = 77) or placebo (n = 80). The average age at screening was 39.2 years, and 23 (15%) were <18 years of age. Of the randomized population, 146 (93%) had confirmed genetic diagnosis of Alport syndrome, and 62% of patients had X-linked mode of inheritance. Mean baseline eGFR was 62.7 mL/min/1.73 m2, and the geometric mean UACR was 141.0 mg/g. The average annual rate of eGFR decline prior to enrollment in the study was -4.9 mL/min/1.73 m2 despite 78% of the patient population receiving ACE inhibitor (ACEi) or ARB therapy. DISCUSSION/CONCLUSION CARDINAL is one of the largest interventional, randomized controlled trials in Alport syndrome conducted to date. Despite the use of ACEi or ARB, patients were experiencing significant loss of kidney function prior to study entry.
Collapse
Affiliation(s)
- Glenn M Chertow
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Palo Alto, California, USA
| | - Gerald B Appel
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Sharon Andreoli
- Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sripal Bangalore
- Cardiovascular Clinical Research Center, New York University School of Medicine, New York, New York, USA
| | - Geoffrey A Block
- Department of Clinical Research and Medical Affairs, US Renal Care, Inc., Plano, Texas, USA
| | - Arlene B Chapman
- Section of Nephrology, University of Chicago, Chicago, Illinois, USA
| | - Melanie P Chin
- Department of Product Development, Reata Pharmaceuticals, Plano, Texas, USA
| | - Keisha L Gibson
- University of North Carolina Kidney Center at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Angie Goldsberry
- Department of Product Development, Reata Pharmaceuticals, Plano, Texas, USA
| | - Kazumoto Iijima
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Lesley A Inker
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts, USA
| | - Bertrand Knebelmann
- Department of Nephrology, Necker Hospital, AP-HP, Université de Paris, Paris, France
| | - Laura H Mariani
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, Michigan, USA
| | - Colin J Meyer
- Department of Product Development, Reata Pharmaceuticals, Plano, Texas, USA
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Megan O'Grady
- Department of Product Development, Reata Pharmaceuticals, Plano, Texas, USA
| | - Arnold L Silva
- Boise Kidney and Hypertension Institute, Meridian, Idaho, USA
| | - Peter Stenvinkel
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, Stockholm, Sweden
| | - Roser Torra
- Inherited Kidney Disorders, Nephrology Department, Fundacio Puigvert, Instituto de Investigacion Carlos III, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Bradley A Warady
- Division of Pediatric Nephrology, Children's Mercy Kansas City, Kansas City, Missouri, USA
| | | |
Collapse
|
30
|
Kashtan CE, Gross O. Clinical practice recommendations for the diagnosis and management of Alport syndrome in children, adolescents, and young adults-an update for 2020. Pediatr Nephrol 2021; 36:711-719. [PMID: 33159213 DOI: 10.1007/s00467-020-04819-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/24/2020] [Accepted: 10/08/2020] [Indexed: 12/28/2022]
Abstract
In 2013, we published a set of clinical practice recommendations for the treatment of Alport syndrome in this journal. We recommended delaying the initiation of angiotensin-converting enzyme inhibition until the onset of overt proteinuria or, in some cases, microalbuminuria. Developments that have occurred over the past 7 years have prompted us to revise these recommendations. We now recommend the initiation of treatment at the time of diagnosis in males with X-linked Alport syndrome and in males and females with autosomal recessive Alport syndrome. We further recommend starting treatment at the onset of microalbuminuria in females with X-linked Alport syndrome and in males and females with autosomal dominant Alport syndrome. This article presents the rationale for these revisions as well as recommendations for diagnostic tactics intended to ensure the early diagnosis of Alport syndrome.
Collapse
Affiliation(s)
- Clifford E Kashtan
- Department of Pediatrics, Division of Pediatric Nephrology, University of Minnesota Medical School, 2450 Riverside Avenue, Minneapolis, MN, 55454, USA.
| | - Oliver Gross
- Department of Nephrology and Rheumatology, University Medical Center Goettingen, Goettingen, Germany
| |
Collapse
|
31
|
Amlie-Wolf L, Baker L, Hiddemen O, Thomas M, Burke C, Gluck C, Zaritsky JJ, Gripp KW. Novel genetic testing model: A collaboration between genetic counselors and nephrology. Am J Med Genet A 2021; 185:1142-1150. [PMID: 33475249 DOI: 10.1002/ajmg.a.62088] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 11/09/2020] [Accepted: 01/09/2021] [Indexed: 11/08/2022]
Abstract
Many barriers to genetic testing currently exist which delay or prevent diagnosis. These barriers include wait times, staffing, education, and cost. Specialists are able to identify patients with disease that may need genetic testing, but lack the genetics support to facilitate that testing in the most cost, time, and medically effective manner. The Nephrology Division and the Genetic Testing Stewardship Program at Nemours A.I. duPont Hospital for Children created a novel service delivery model in which nephrologists and genetic counselors collaborate in order to highlight their complementary strengths (clinical expertise of nephrologists and genetics and counseling skills of genetic counselors). This collaboration has reduced many barriers to care for our patients. This workflow facilitated the offering of genetic testing to 76 patients, with 86 tests completed over a 20-month period. Thirty-two tests were deferred. Twenty-seven patients received a diagnosis, which lead to a change in their medical management, three of whom were diagnosed by cascade family testing. Forty-two patients had a negative result and 16 patients had one or more variants of uncertain significance on testing. The inclusion of genetic counselors in the workflow is integral toward choosing the most cost and time effective genetic testing strategy, as well as providing psychosocial support to families. The genetic counselors obtain informed consent, and review genetic test results and recommendations with the patient and their family. The availability of this program to our patients increased access to genetic testing and helps to provide diagnoses and supportive care.
Collapse
Affiliation(s)
- Louise Amlie-Wolf
- Precision Medicine/Genetic Testing Stewardship Program, Nemours A.I. duPont Hospital for Children Precision Medicine/Genetic Testing Stewardship Program, Wilmington, Delaware, USA
| | - Laura Baker
- Precision Medicine/Genetic Testing Stewardship Program, Nemours A.I. duPont Hospital for Children Precision Medicine/Genetic Testing Stewardship Program, Wilmington, Delaware, USA
| | - Olivia Hiddemen
- Precision Medicine/Genetic Testing Stewardship Program, Nemours A.I. duPont Hospital for Children Precision Medicine/Genetic Testing Stewardship Program, Wilmington, Delaware, USA
| | - Morgan Thomas
- Precision Medicine/Genetic Testing Stewardship Program, Nemours A.I. duPont Hospital for Children Precision Medicine/Genetic Testing Stewardship Program, Wilmington, Delaware, USA
| | - Christine Burke
- Division of Nephrology, Nemours A.I. duPont Hospital for Children, Wilmington, Delaware, USA
| | - Caroline Gluck
- Division of Nephrology, Nemours A.I. duPont Hospital for Children, Wilmington, Delaware, USA
| | - Joshua J Zaritsky
- Division of Nephrology, Nemours A.I. duPont Hospital for Children, Wilmington, Delaware, USA
| | - Karen W Gripp
- Precision Medicine/Genetic Testing Stewardship Program, Nemours A.I. duPont Hospital for Children Precision Medicine/Genetic Testing Stewardship Program, Wilmington, Delaware, USA
| |
Collapse
|
32
|
Lodeweyckx N, Wouters K, Ledeganck KJ, Trouet D. Biopsy or Biomarker? Children With Minimal Change Disease Have a Distinct Profile of Urinary Epidermal Growth Factor. Front Pediatr 2021; 9:727954. [PMID: 34900856 PMCID: PMC8657767 DOI: 10.3389/fped.2021.727954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 10/05/2021] [Indexed: 02/02/2023] Open
Abstract
Background: In this study, the profile of urinary EGF excretion (uEGF/uCreat) was mapped in children presenting with prolonged proteinuria or with nephrotic syndrome refractory to or dependent of steroids. We investigated whether uEGF/uCreat could be linked to the underlying biopsy result, taking into account its response to immunosuppressive medication and to ACE inhibition, as well as genetic predisposition. Methods: Ninety-eight pediatric patients with initial presentation of nephrotic syndrome or prolonged proteinuria were included in this study, along with 49 healthy controls and 20 pediatric Alport patients. All patients had a normal kidney function and were normotensive during the course of the study, whether or not under ACE inhibition. In repeated urine samples, uEGF was measured and concentration was normalized by urine creatinine. In order to compare diagnosis on kidney biopsy, genetic predisposition and response of uEGF/uCreat to immunosuppression and to ACE inhibition, uEGF/uCreat is studied in a linear mixed effects model. Results: Patients with Minimal Change Disease (MCD) showed a significantly different profile of uEGF/uCreat in comparison to healthy children, as well as compared to patients with Focal Segmental Glomerulosclerosis (FSGS) or another glomerulopathy on kidney biopsy. The response of uEGF/uCreat to ACE inhibition was absent in minimal change disease and contrasted with an impressive beneficial effect of ACE inhibition on uEGF/uCreat in FSGS and other proteinuric glomerulopathies. Absence of a genetic predisposition was also associated with a significantly lower uEGF/uCreat. Conclusions: Despite preserved kidney function, children with a proteinuric or nephrotic glomerular disease on kidney biopsy show a significantly lower uEGF/uCreat, indicative of early tubulo-interstitial damage, which appears reversible under ACE inhibition in any underlying glomerulopathy except in minimal change disease. In view of the distinct profile of uEGF/uCreat in minimal change disease compared to other glomerulopathies, and the link between genetic predisposition and uEGF/uCreat, our study suggests that uEGF/uCreat can be a helpful tool to decide on the need for a renal biopsy in order to differentiate minimal change disease from other proteinuric glomerular diseases.
Collapse
Affiliation(s)
- Niels Lodeweyckx
- Department of Pediatric Nephrology, Antwerp University Hospital, Edegem, Belgium
| | - Kristien Wouters
- Clinical Trial Center (CTC), Clinical Research Center (CRC) Antwerp, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
| | - Kristien J Ledeganck
- Laboratory of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, University of Antwerp, Edegem, Belgium
| | - Dominique Trouet
- Department of Pediatric Nephrology, Antwerp University Hospital, Edegem, Belgium.,Laboratory of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, University of Antwerp, Edegem, Belgium
| |
Collapse
|
33
|
Genotype-phenotype correlations and nephroprotective effects of RAAS inhibition in patients with autosomal recessive Alport syndrome. Pediatr Nephrol 2021; 36:2719-2730. [PMID: 33772369 PMCID: PMC8370956 DOI: 10.1007/s00467-021-05040-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 02/20/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Autosomal recessive Alport syndrome (ARAS) is caused by pathogenic variants in both alleles of either COL4A3 or COL4A4 genes. Reports on ARAS are rare due to small patient numbers and there are no reports on renin-angiotensin-aldosterone system (RAAS) inhibition therapy in ARAS. METHODS Retrospective study in 101 patients with ARAS from Chinese Registry Database of Hereditary Kidney Diseases and European Alport Registry. Genotype-phenotype correlations and nephroprotective effects of RAAS inhibition in ARAS were evaluated. RESULTS Median age was 15 years (range 1.5-46 years). Twelve patients progressed to stage 5 chronic kidney disease (CKD5) at median age 20.5 years. Patients without missense variants had both higher prevalence and earlier onset age of hearing loss, nephrotic-range proteinuria, more rapid decline of eGFR, and earlier onset age of CKD5 compared to patients with 1 or 2 missense variants. Most patients (79/101, 78%) currently are treated with RAAS inhibitors; median age at therapy initiation was 10 years and mean duration 6.5 ± 6.0 years. Median age at CKD5 for untreated patients was 24 years. RAAS inhibition therapy delayed CKD5 onset in those with impaired kidney function (T-III) to median age 35 years, but is undefined in treated patients with proteinuria (T-II) due to low number of events. No treated patients with microalbuminuria (T-I) progressed to CKD5. ARAS patients with 1 or 2 missense variants showed better response to treatment than patients with non-missense-variants. CONCLUSIONS Our study provides the first evidence for early use of RAAS inhibition therapy in patients with ARAS. Furthermore, genotype in ARAS correlates with response to therapy in favor of missense variants.
Collapse
|
34
|
Mastrangelo A, Giani M, Groppali E, Castorina P, Soldà G, Robusto M, Fallerini C, Bruttini M, Renieri A, Montini G. X-Linked Alport Syndrome in Women: Genotype and Clinical Course in 24 Cases. Front Med (Lausanne) 2020; 7:580376. [PMID: 33330536 PMCID: PMC7719790 DOI: 10.3389/fmed.2020.580376] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/19/2020] [Indexed: 11/30/2022] Open
Abstract
Objectives: X-linked Alport syndrome (XLAS) females are at risk of developing proteinuria and chronic kidney damage (CKD). The aim of this study is to evaluate the genotype-phenotype correlation in this rare population. Materials and Methods: This is a prospective, observational study of XLAS females, confirmed by a pathogenic mutation in COL4A5 and renal ultrastructural evaluation. Proteinuria, renal function and extrarenal involvement were monitored during follow-up. Patients were divided in 2 groups, according to mutations in COL4A5: missense (Group 1) and non-missense variants (Group 2). Results: Twenty-four XLAS females, aged 10.6 ± 10.4 years at clinical onset (mean follow-up: 13.1 ± 12.6 years) were recruited between 2000 and 2017 at a single center. In group 1 there were 10 patients and in group 2, 14 (mean age at the end of follow-up: 24.9 ± 13.6 and 23.2 ± 13.8 years, respectively). One patient in Group 1 and 9 in Group 2 (p = 0.013) developed proteinuria during follow-up. Mean eGFR at last follow-up was lower in Group 2 (p = 0.027), where two patients developed CKD. No differences in hearing loss were documented among the two groups. Two patients in Group 2 carried one mutation in both COL4A5 and COL4A3 (digenic inheritance) and were proteinuric. In one family, the mother presented only hematuria while the daughter was proteinuric and presented a greater inactivation of the X chromosome carrying the wild-type allele. Conclusions: The appearance of proteinuria and CKD is more frequent in patients with severe variants. Carrying digenic inheritance and skewed XCI seem to be additional risk factors for proteinuria in XLAS females.
Collapse
Affiliation(s)
- Antonio Mastrangelo
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marisa Giani
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elena Groppali
- Department of Pediatrics, V. Buzzi Children's Hospital, Milan, Italy
| | | | - Giulia Soldà
- Dipartimento di Scienze Biomediche, Humanitas University, Milan, Italy.,Humanitas Clinical and Research Center, Milan, Italy
| | - Michela Robusto
- Experimental Therapeutics Program, Istituto FIRC di Oncologia Molecolare-Fondazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology Foundation, Milan, Italy
| | | | - Mirella Bruttini
- Azienda Ospedaliera Universitaria Senese, Medical Genetics, University of Siena, Siena, Italy
| | - Alessandra Renieri
- Azienda Ospedaliera Universitaria Senese, Medical Genetics, University of Siena, Siena, Italy
| | - Giovanni Montini
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Giuliana and Bernardo Caprotti Chair of Pediatrics, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
35
|
Angiotensin-converting enzyme inhibitors in patients with Alport syndrome: can all patients benefit? Kidney Int 2020; 98:1400-1402. [PMID: 33276866 DOI: 10.1016/j.kint.2020.07.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 12/22/2022]
Abstract
Angiotensin-converting enzyme inhibitors (ACEi) and angiotensin receptor blockers (ARBs) are prescribed to slow the progression of kidney disease in patients with Alport syndrome. In a recent publication by Yamamura et al. the authors showed an association of ACEi or ARB treatment with delay in ESKD, even for those patients with severe, truncating mutations. Despite these encouraging findings, there remain a number of clinical questions about the use of ACEi and ARBs in Alport syndrome.
Collapse
|
36
|
Ozdemir G, Gulhan B, Atayar E, Saygılı S, Soylemezoglu O, Ozcakar ZB, Eroglu FK, Candan C, Demir BK, Soylu A, Yüksel S, Alpay H, Agbas A, Duzova A, Hayran M, Ozaltin F, Topaloglu R. COL4A3 mutation is an independent risk factor for poor prognosis in children with Alport syndrome. Pediatr Nephrol 2020; 35:1941-1952. [PMID: 32394188 DOI: 10.1007/s00467-020-04574-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/03/2020] [Accepted: 04/07/2020] [Indexed: 01/14/2023]
Abstract
BACKGROUND Alport syndrome (AS) is an inherited glomerular disease caused by mutations in COL4A3, COL4A4, or COL4A5. Associations between clinical manifestations and genotype are not yet well defined. Our study aimed to define clinical and genetic characteristics, establish genotype-phenotype correlations, and determine prognosis of AS in children. METHODS A total of 87 children with AS from 10 pediatric nephrology centers, whom had genetic analyses performed at the Hacettepe University Nephrogenetics Laboratory between February 2017 and February 2019, were included. Data regarding demographics, family history, clinical and laboratory characteristics, histopathological and genetic test results, treatments, and yearly follow-up results were retrospectively analyzed. RESULTS Of 87 patients, 16% presented with nephrotic syndrome. In patients with nephrotic syndrome, kidney biopsy findings showed focal segmental glomerulosclerosis (FSGS) in 79%, and COL4A3 mutations were the leading genetic abnormality (50%). Twenty-four percent of all patients progressed to chronic kidney disease (CKD). The rate of progression to CKD and the decline in the glomerular filtration rate of the patients with COL4A3 mutation were higher than other mutation groups (p < 0.001 and p = 0.04, respectively). In kidney survival analysis, nephrotic syndrome presentation, histopathology of FSGS, COL4A3 mutations, and autosomal recessive inheritance were found as independent risk factors for earlier progression to CKD. Cyclosporin A treatment did not improve kidney survival. CONCLUSIONS We emphasize that genetic testing is important for patients suspected as having AS. Furthermore, COL4A mutations should be considered in patients with FSGS and steroid-resistant nephrotic syndrome. This approach will shed light on the prognosis of patients and help with definitive diagnosis, preventing unnecessary and potentially harmful medications. Graphical abstract.
Collapse
MESH Headings
- Adolescent
- Autoantigens/genetics
- Biopsy
- Child
- Child, Preschool
- Collagen Type IV/genetics
- DNA Mutational Analysis
- Disease Progression
- Female
- Follow-Up Studies
- Genetic Association Studies
- Genetic Testing
- Glomerulosclerosis, Focal Segmental/diagnosis
- Glomerulosclerosis, Focal Segmental/epidemiology
- Glomerulosclerosis, Focal Segmental/genetics
- Glomerulosclerosis, Focal Segmental/pathology
- Humans
- Kidney/pathology
- Male
- Mutation
- Nephritis, Hereditary/complications
- Nephritis, Hereditary/diagnosis
- Nephritis, Hereditary/genetics
- Nephritis, Hereditary/pathology
- Prognosis
- Renal Insufficiency, Chronic/diagnosis
- Renal Insufficiency, Chronic/epidemiology
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/pathology
- Retrospective Studies
Collapse
Affiliation(s)
- Gulsah Ozdemir
- Division of Pediatric Nephrology, Faculty of Medicine, Hacettepe University, 06230, Ankara, Turkey
| | - Bora Gulhan
- Division of Pediatric Nephrology, Faculty of Medicine, Hacettepe University, 06230, Ankara, Turkey
| | - Emine Atayar
- Division of Pediatric Nephrology, Nephrogenetics Laboratory, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Seha Saygılı
- Division of Pediatric Nephrology, Faculty of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Oguz Soylemezoglu
- Division of Pediatric Nephrology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Zeynep Birsin Ozcakar
- Division of Pediatric Nephrology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Fehime Kara Eroglu
- Division of Pediatric Nephrology, Dr. Sami Ulus Maternity and Children's Health Hospital, Ankara, Turkey
| | - Cengiz Candan
- Division of Pediatric Nephrology, Göztepe Training and Research Hospital, Istanbul Medeniyet University, Istanbul, Turkey
| | - Belde Kasap Demir
- Division of Pediatric Nephrology, Tepecik Training and Research Hospital, Izmir, Turkey
| | - Alper Soylu
- Division of Pediatric Nephrology, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey
| | - Selçuk Yüksel
- Division of Pediatric Nephrology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Harika Alpay
- Division of Pediatric Nephrology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Ayse Agbas
- Division of Pediatric Nephrology, Haseki Training and Research Hospital, Istanbul, Turkey
| | - Ali Duzova
- Division of Pediatric Nephrology, Faculty of Medicine, Hacettepe University, 06230, Ankara, Turkey
| | - Mutlu Hayran
- Department of Preventive Oncology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Fatih Ozaltin
- Division of Pediatric Nephrology, Faculty of Medicine, Hacettepe University, 06230, Ankara, Turkey
- Division of Pediatric Nephrology, Nephrogenetics Laboratory, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Rezan Topaloglu
- Division of Pediatric Nephrology, Faculty of Medicine, Hacettepe University, 06230, Ankara, Turkey.
| |
Collapse
|
37
|
Nicolaou O, Kousios A, Sokratous K, Potamiti L, Koniali L, Neophytou G, Papacharalampous R, Zanti M, Ioannou K, Hadjisavvas A, Stingl C, Luider TM, Kyriacou K. Alport syndrome: Proteomic analysis identifies early molecular pathway alterations in Col4a3 knock out mice. Nephrology (Carlton) 2020; 25:937-949. [PMID: 32743880 PMCID: PMC7754404 DOI: 10.1111/nep.13764] [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: 03/28/2020] [Revised: 07/06/2020] [Accepted: 07/26/2020] [Indexed: 01/02/2023]
Abstract
AIM Alport syndrome (AS) is the second most common hereditary kidney disease caused by mutations in collagen IV genes. Patients present with microhaematuria that progressively leads to proteinuria and end stage renal disease. Currently, no specific treatment exists for AS. Using mass spectrometry based proteomics, we aimed to detect early alterations in molecular pathways implicated in AS before the stage of overt proteinuria, which could be amenable to therapeutic intervention. METHODS Kidneys were harvested from male Col4a3-/- knock out and sex and age-matched Col4a3+/+ wild-type mice at 4 weeks of age. Purified peptides were separated by liquid chromatography and analysed by high resolution mass spectrometry. The Cytoscape bioinformatics tool was used for function enrichment and pathway analysis. PPARα expression levels were evaluated by immunofluorescence and immunoblotting. RESULTS Proteomic analysis identified 415 significantly differentially expressed proteins, which were mainly involved in metabolic and cellular processes, the extracellular matrix, binding and catalytic activity. Pathway enrichment analysis revealed among others, downregulation of the proteasome and PPAR pathways. PPARα protein expression levels were observed to be downregulated in Alport mice, supporting further the results of the discovery proteomics. CONCLUSION This study provides additional evidence that alterations in proteins which participate in cellular metabolism and mitochondrial homeostasis in kidney cells are early events in the development of chronic kidney disease in AS. Of note is the dysregulation of the PPAR pathway, which is amenable to therapeutic intervention and provides a new potential target for therapy in AS.
Collapse
Affiliation(s)
- Orthodoxia Nicolaou
- Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.,Department of Electron Microscopy and Molecular Pathology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Andreas Kousios
- Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.,Renal and Transplant Centre, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Kleitos Sokratous
- Department of Electron Microscopy and Molecular Pathology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Louiza Potamiti
- Department of Electron Microscopy and Molecular Pathology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Lola Koniali
- Department of Molecular Genetics Thalassemia, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - George Neophytou
- Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Revekka Papacharalampous
- Department of Neurology Clinic A, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Maria Zanti
- Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.,Department of Electron Microscopy and Molecular Pathology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.,Bioinformatics Group, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Kyriakos Ioannou
- Department of Nephrology, Apollonion Private Hospital, Nicosia, Cyprus.,School of Medicine, European University, Nicosia, Cyprus
| | - Andreas Hadjisavvas
- Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.,Department of Electron Microscopy and Molecular Pathology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Christoph Stingl
- Laboratory of Neuro-Oncology, Clinical and Cancer Proteomics, Department of Neurology, Erasmus University Medical Center Rotterdam, Rotterdam, CN, The Netherlands
| | - Theo M Luider
- Laboratory of Neuro-Oncology, Clinical and Cancer Proteomics, Department of Neurology, Erasmus University Medical Center Rotterdam, Rotterdam, CN, The Netherlands
| | - Kyriacos Kyriacou
- Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.,Department of Electron Microscopy and Molecular Pathology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| |
Collapse
|
38
|
Kashtan CE. An update on current and potential genetic insights and diagnosis of Alport syndrome. Expert Opin Orphan Drugs 2020. [DOI: 10.1080/21678707.2020.1784722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
39
|
Kashtan CE. Alport Syndrome: Achieving Early Diagnosis and Treatment. Am J Kidney Dis 2020; 77:272-279. [PMID: 32712016 DOI: 10.1053/j.ajkd.2020.03.026] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/24/2020] [Indexed: 12/11/2022]
Abstract
Alport syndrome is a genetically and phenotypically heterogeneous disorder of glomerular, cochlear, and ocular basement membranes resulting from mutations in the collagen IV genes COL4A3, COL4A4, and COL4A5. Alport syndrome can be transmitted as an X-linked, autosomal recessive, or autosomal dominant disorder. Individuals with Alport syndrome have a significant lifetime risk for kidney failure, as well as sensorineural deafness and ocular abnormalities. The availability of effective intervention for Alport syndrome-related kidney disease makes early diagnosis crucial, but this can be impeded by the genotypic and phenotypic complexity of the disorder. This review presents an approach to enhancing early diagnosis and achieving optimal outcomes.
Collapse
Affiliation(s)
- Clifford E Kashtan
- Pediatric Nephrology, University of Minnesota Medical School, Minneapolis, MN.
| |
Collapse
|
40
|
Wang C, Liang S, Xing S, Xu K, Xiao H, Deng H, Wang X, Chen L, Ding J, Wang F. Endoplasmic Reticulum Stress Activation in Alport Syndrome Varies Between Genotype and Cell Type. Front Genet 2020; 11:36. [PMID: 32117450 PMCID: PMC7025644 DOI: 10.3389/fgene.2020.00036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/13/2020] [Indexed: 12/26/2022] Open
Abstract
Alport syndrome is a hereditary progressive chronic kidney disease caused by mutations in type IV collagen genes COL4A3/4/5. X-linked Alport syndrome (XLAS) is caused by mutations in the COL4A5 gene and is the most common form of Alport syndrome. A strong correlation between the type of COL4A5 mutation and the age developing end-stage renal disease in male patients has been found. Mutation to the α (IV) chain causes retention of the protein to the endoplasmic reticulum lumen, which causes endoplasmic reticulum stress (ERS) and subsequent exertion of deleterious intracellular effects through the activation of ERS. The exact time point that mutant type IV collagen α chain exerts its deleterious effects remains elusive. In this study, we explored the relationship between the COL4A5 genotype and cell type in ERS activation. We obtained skin fibroblasts from Alport syndrome patients with different COL4A5 mutation categories [i.e., a missense mutation (c.4298G > T, p.Gly1433Val) in exon 47, a splicing mutation (c.1949–1G > A) in intron 25 and an insertion (c.573_c.574insG, p. Pro193Alafs*23) in exon 10], and then reprogrammed these fibroblasts into induced pluripotent stem cells (iPSCs). Interestingly, no significant dysregulation of ERS pathway markers was observed for the three COL4A5 mutant iPSCs; however, significant activation of ERS in COL4A5 mutant fibroblasts was observed. In addition, we found that the activation levels of some ERS markers in fibroblasts varied among the three COL4A5 mutation types. Mutant COL4A5 proteins were demonstrated to have different effects on cells at different stages of ontogenesis, providing a theoretical basis for choosing the timing of intervention. The observed differences in activation of ERS by the COL4A5 mutant fibroblasts may contribute to the intracellular molecular mechanisms that describe the correlation between genotype and clinical features in XLAS.
Collapse
Affiliation(s)
- Cong Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Shenghui Liang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Shijia Xing
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Ke Xu
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Huijie Xiao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Haiyue Deng
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Xiaoyuan Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Liangyi Chen
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Jie Ding
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Fang Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| |
Collapse
|
41
|
A multicenter, randomized, placebo-controlled, double-blind phase 3 trial with open-arm comparison indicates safety and efficacy of nephroprotective therapy with ramipril in children with Alport's syndrome. Kidney Int 2020; 97:1275-1286. [PMID: 32299679 DOI: 10.1016/j.kint.2019.12.015] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/05/2019] [Accepted: 12/13/2019] [Indexed: 01/09/2023]
Abstract
Children with Alport syndrome develop renal failure early in life. Since the safety and efficacy of preemptive nephroprotective therapy are uncertain we conducted a randomized, placebo-controlled, double-blind trial in 14 German sites of pediatric patients with ramipril for three to six years plus six months follow-up to determine these parameters. Pretreated children and those whose parents refused randomization became an open-arm control, which were compared to prospective real-world data from untreated children. The co-primary endpoints were safety (adverse drug reactions) and efficacy (time to progression). Out of 66 oligosymptomatic children, 22 were randomized and 44 joined the open-arm comparison. Ramipril therapy showed no safety issues (total of 216.4 patient-years on ramipril; adverse event rate-ratio 1.00; 95% confidence interval 0.66-1.53). Although not significant, our results cautiously showed that ramipril therapy was effective: in the randomized arm, Ramipril decreased the risk of disease progression by almost half (hazard ratio 0.51 (0.12-2.20)), diminished the slope of albuminuria progression and the decline in glomerular filtration. In adjusted analysis, indications of efficacy were supported by prospective data from participants treated open label compared with untreated children, in whom ramipril again seemed to reduce progression by almost half (0.53 (0.22-1.29)). Incorporating these results into the randomized data by Bayesian evidence synthesis resulted in a more precise estimate of the hazard-ratio of 0.52 (0.19-1.39). Thus, our study shows the safety of early initiation of therapy and supports the hope to slow renal failure by many years, emphasizing the value of preemptive therapy. Hence, screening programs for glomerular hematuria in children and young adults could benefit from inclusion of genetic testing for Alport-related gene-variants.
Collapse
|
42
|
Rheault MN, Savige J, Randles MJ, Weinstock A, Stepney M, Turner AN, Parziale G, Gross O, Flinter FA, Miner JH, Lagas S, Gear S, Lennon R. The importance of clinician, patient and researcher collaborations in Alport syndrome. Pediatr Nephrol 2020; 35:733-742. [PMID: 31044288 PMCID: PMC7096363 DOI: 10.1007/s00467-019-04241-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 03/15/2019] [Indexed: 12/19/2022]
Abstract
Alport syndrome is caused by mutations in the genes COL4A3, COL4A4 or COL4A5 and is characterised by progressive glomerular disease, sensorineural hearing loss and ocular defects. Occurring in less than 1:5000, Alport syndrome is a rare genetic disorder but still accounts for > 1% of the prevalent population receiving renal replacement therapy. There is also increasing awareness about the risk of chronic kidney disease in individuals with heterozygous mutations in Alport syndrome genes. The mainstay of current therapy is the use of angiotensin-converting enzyme inhibitors and angiotensin receptor blockers, yet potential new therapies are now entering clinical trials. The 2017 International Workshop on Alport Syndrome in Glasgow was a pre-conference workshop ahead of the 50th anniversary meeting of the European Society for Pediatric Nephrology. It focussed on updates in clinical practice, genetics and basic science and also incorporated patient perspectives. More than 80 international experts including clinicians, geneticists, researchers from academia and industry, and patient representatives took part in panel discussions and breakout groups. This report summarises the workshop proceedings and the relevant contemporary literature. It highlights the unique clinician, patient and researcher collaborations achieved by regular engagement between the groups.
Collapse
Affiliation(s)
- Michelle N. Rheault
- Department of Pediatrics, University of Minnesota Masonic Children’s Hospital, Minneapolis, MN USA
| | - Judith Savige
- Department of Medicine, Royal Melbourne Hospital, Victoria, Australia
| | - Michael J. Randles
- Wellcome Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | | | - Melissa Stepney
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - A Neil Turner
- Renal Medicine, Royal Infirmary, University of Edinburgh, Edinburgh, UK
| | | | - Oliver Gross
- Clinic of Nephrology and Rheumatology, University Medicine Goettingen, Goettingen, Germany
| | - Frances A Flinter
- Department of Clinical Genetics, Guy’s & St Thomas’ NHS Foundation Trust, London, UK
| | - Jeffrey H Miner
- Division of Nephrology, Washington University School of Medicine, St Louis, MO USA
| | | | | | - 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, University of Manchester, Manchester, UK. .,Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
43
|
CUBN variants uncouple proteinuria from kidney function. Nat Rev Nephrol 2019; 16:135-136. [PMID: 31836876 DOI: 10.1038/s41581-019-0242-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
44
|
Moshirfar M, Skanchy DF, Gomez AT, Ronquillo YC, Buckner B, Hoopes PC. Keratoconus in a patient with Alport syndrome: A case report. World J Clin Cases 2019; 7:3012-3017. [PMID: 31624748 PMCID: PMC6795741 DOI: 10.12998/wjcc.v7.i19.3012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 08/19/2019] [Accepted: 09/09/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Known ocular manifestations of Alport syndrome include features such as anterior lenticonus and fleck retinopathy. Reports of keratoconus in such patients are limited. We report tomographic findings consistent with keratoconus in a patient with Alport syndrome.
CASE SUMMARY A 52-year-old female was referred to our ophthalmology clinic with decreased vision and increased tearing. She was diagnosed with stage III Alport syndrome two years prior. Upon examination she was found to have average keratometries of 48 D bilaterally with tomographic evidence of keratoconus.
CONCLUSION Although a rare presentation, concurrent Alport syndrome and keratoconus should be considered when reviewing the ocular health of Alport syndrome patients and appropriate management steps should be taken upon the diagnosis.
Collapse
Affiliation(s)
- Majid Moshirfar
- John A. Moran Eye Center, Department of Ophthalmology and Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT 84101, United States
- Utah Lions Eye Bank, Salt Lake City, UT 84101, United States
- Hoopes Durrie Rivera Research Center, Hoopes Vision, Draper, UT 84020, United States
| | - David F Skanchy
- Kellogg Eye Center, Department of Ophthalmology, University of Michigan Medical School, Ann Arbor, MI 48103, United States
| | - Aaron T Gomez
- The University of Texas Rio Grande Valley School of Medicine, Edinburg, TX 78542, United States
| | - Yasmyne C Ronquillo
- Hoopes Durrie Rivera Research Center, Hoopes Vision, Draper, UT 84020, United States
| | - Benjamin Buckner
- Hoopes Durrie Rivera Research Center, Hoopes Vision, Draper, UT 84020, United States
| | - Phillip C Hoopes
- Hoopes Durrie Rivera Research Center, Hoopes Vision, Draper, UT 84020, United States
| |
Collapse
|
45
|
Zhang Y, Ding J, Wang S, Zhang H, Zhong X, Liu X, Xu K, Wang F. Reassessing the pathogenicity of c.2858G>T(p.(G953V)) in COL4A5 Gene: report of 19 Chinese families. Eur J Hum Genet 2019; 28:244-252. [PMID: 31576025 PMCID: PMC6974604 DOI: 10.1038/s41431-019-0523-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 09/03/2019] [Accepted: 09/17/2019] [Indexed: 11/09/2022] Open
Abstract
X-linked Alport syndrome (XLAS) is an inherited renal disease caused by mutations in COL4A5 gene. The c.2858G>T(p.(G953V)) in COL4A5 gene (rs78972735) has been considered pathogenic previously. However, there are conflicting interpretations of its pathogenicity recently. Here we presented 19 Chinese families, out of which 36 individuals (18 probands and 18 family members) carried the c.2858G>T(p.(G953V)) in COL4A5 gene. The clinical manifestations and genetic findings of them were analyzed. We found there were no clinical features of Alport syndrome not only in six probands with c.2858G>T(p.(G953V)) in COL4A5 plus pathogenic variants in other genes (e.g., WT1, ADCK4, NPHP1, TRPC6, COL4A4, and PAX2) but also in another six probands with only the c.2858G>T(p.(G953V)) variant. The other six probands with a combination of c.2858G>T(p.(G953V)) and another pathogenic variant in COL4A5 had XLAS. Eleven family members (11/18, nine females and two males) who had only the c.2858G>T(p.(G953V)) variant were asymptomatic. These two males (at age of 42 and 35 years) had normal result of urine analysis and no more clinical traits of Alport syndrome. We conclude c.2858G>T(p.(G953V)) in COL4A5 gene is not a pathogenic variant for XLAS. Individuals should not be diagnosed as XLAS only based on the detection of c.2858G>T(p.(G953V)) in COL4A5 gene.
Collapse
Affiliation(s)
- Yanqin Zhang
- Department of Pediatrics, Peking University First Hospital, 100034, Beijing, China
| | - Jie Ding
- Department of Pediatrics, Peking University First Hospital, 100034, Beijing, China
| | - Suxia Wang
- Department of Electron Microscopy, Peking University First Hospital, 100034, Beijing, China
| | - Hongwen Zhang
- Department of Pediatrics, Peking University First Hospital, 100034, Beijing, China
| | - Xuhui Zhong
- Department of Pediatrics, Peking University First Hospital, 100034, Beijing, China
| | - Xiaoyu Liu
- Department of Pediatrics, Peking University First Hospital, 100034, Beijing, China
| | - Ke Xu
- Department of Pediatrics, Peking University First Hospital, 100034, Beijing, China
| | - Fang Wang
- Department of Pediatrics, Peking University First Hospital, 100034, Beijing, China.
| |
Collapse
|
46
|
Sun H, Yu X, Li S, Xu H, Yang J, Yi T, Han P, Shao M. A novel mutation in a Chinese family with autosomal recessive Alport syndrome: a case report. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:3565-3569. [PMID: 31934206 PMCID: PMC6949818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/26/2019] [Indexed: 06/10/2023]
Abstract
Alport syndrome (AS) is a familial hereditary nephropathy which is characterized by molecular abnormalities in Collagen IV a345. As more gene mutations are discovered, it has been reported that autosomal recessive disease accounts for a smaller proportion (about 4%) of AS patients than previously recognized. We report here a novel mutation in COL4A4 in a Chinese family with autosomal recessive AS. Patient 1 was a 24-year-old Chinese man. He and his brother (patient 2) had a history of proteinuria and hematuria with renal dysfunction and sensorineural deafness. Pathologic findings were consistent with Alport syndrome, and genetic analysis revealed that both patients had two heterozygous mutations, c.1423 G>T (p.Gly475Cys) in EX21/CDS20 and c.735 G>A (p.Pro245Pro) in EX12/CDS11, and that each mutation originated from their mother or father who were carriers for one of these two mutations. Both patients showed similar results by laboratory examination and histopathologic assessment. Patient 1 received ACEI treatment and ran a stable clinical course, whereas patient 2 refused ACEI treatment and had progressive deterioration of renal function. This is the first report of a novel mutation in the collagen domain of COL4A4 gene. The results add to the spectrum of mutations in COL4A4 of Alport syndrome.
Collapse
Affiliation(s)
- Huili Sun
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen 518033, Guangdong, China
| | - Xuewen Yu
- Department of Pathology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen 518033, Guangdong, China
| | - Shunmin Li
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen 518033, Guangdong, China
| | - Hua Xu
- Department of Pathology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen 518033, Guangdong, China
| | - Jun Yang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen 518033, Guangdong, China
| | - Tiegang Yi
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen 518033, Guangdong, China
| | - Pengxun Han
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen 518033, Guangdong, China
| | - Mumin Shao
- Department of Pathology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen 518033, Guangdong, China
| |
Collapse
|
47
|
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
|
48
|
YE Q, ZHANG Y, WANG J, MAO J. [Analysis of Alport syndrome induced by type IV collagen alpha 5 gene mutation in two families]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2019; 48:384-389. [PMID: 31901041 PMCID: PMC8800710 DOI: 10.3785/j.issn.1008-9292.2019.08.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 08/14/2019] [Indexed: 06/10/2023]
Abstract
OBJECTIVE To investigate genetic characteristics of Alport syndrome. METHODS High-throughput sequencing-based whole exome sequencing was performed in two patients with recurrent unexplained abnormal urinalysis. The pathogenicity of the genetic variations, type of Mendelian genetics, and clinical phenotypes were analysed, and the disease-cause mutations were confirmed in the family members using Sanger sequencing. RESULTS Two heterozygous splice site mutations of COL4A5 gene c.2147-2A > T (IVS27) and c.646-2A > G (IVS11) (NM_033380) were found in patients of the two families, which showed a co-segregation association with the affected members of the families. CONCLUSIONS Alport syndrome is mainly inherited from direct female patients, and prenatal genetic screening based on amniotic fluid testing can effectively prevent birth defects in patients with a family history of this characteristic phenotype.
Collapse
Affiliation(s)
| | | | | | - Jianhua MAO
- 毛建华(1969—), 男, 博士, 主任医师, 博士生导师, 主要从事小儿肾脏疾病研究; E-mail:
;
https://orcid.org/0000-0002-6076-3806
| |
Collapse
|
49
|
Nephroprotektive Therapie am Beispiel des Alport-Syndroms. Monatsschr Kinderheilkd 2019. [DOI: 10.1007/s00112-018-0641-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
50
|
Zhang Y, Ding J, Zhang H, Yao Y, Xiao H, Wang S, Wang F. Effect of heterozygous pathogenic COL4A3 or COL4A4 variants on patients with X-linked Alport syndrome. Mol Genet Genomic Med 2019; 7:e647. [PMID: 30883042 PMCID: PMC6503168 DOI: 10.1002/mgg3.647] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/03/2019] [Accepted: 02/26/2019] [Indexed: 12/12/2022] Open
Abstract
Background Alport syndrome is an inherited renal disease caused by mutations in COL4A3, COL4A4, or COL4A5 genes. Coexisting mutations in either two of the three genes in Alport patients have been reported recently. However, the effect of heterozygous mutations in COL4A3 or COL4A4 genes in X‐linked Alport syndrome (XLAS) patients is unclear. Methods Using targeted next‐generation sequencing, six unrelated Chinese children were identified to have a combination of a pathogenic variant in COL4A5 and a heterozygous mutation in COL4A3 or COL4A4. They were three males and three females. Another three XLAS males each with only one pathogenic variant in COL4A5 were included. The clinical data were analyzed and compared between the males in two groups (group 1, males with a pathogenic variant in COL4A5 and a heterozygous pathogenic variant in COL4A3 or COL4A4; group 2, males with only one pathogenic variant in COL4A5). Results Patients with XLAS who also had heterozygous pathogenic COL4A3 or COL4A4 variants accounted for 1% of Alport syndrome. In this study, three children showed coexisting pathogenic variants in COL4A5 and COL4A3. Two children showed pathogenic variants in COL4A5 and COL4A4. One child had pathogenic variants in the three COL4A3‐5 genes, in which the pathogenic variant in COL4A5 was de novo and the pathogenic variants in COL4A4 and COL4A3 were inherited independently (in trans). The site and type of mutations in COL4A5 were similar between the two groups. It was revealed that males in group 1 presented more severe proteinuria than males in group 2 (p < 0.05). Conclusion The present study provides further evidence for complicated genotype in Alport syndrome. For the first time, we reported a case with three pathogenic variants in COL4A5, COL4A3, and COL4A4 genes. Moreover, we found that heterozygous pathogenic COL4A3 or COL4A4 variants are likely to make XLAS disease more serious.
Collapse
Affiliation(s)
- Yanqin Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Jie Ding
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Hongwen Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yong Yao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Huijie Xiao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Suxia Wang
- Department of Electron Microscopy, Peking University First Hospital, Beijing, China
| | - Fang Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| |
Collapse
|