Skip to main content
Erschienen in: BMC Nephrology 1/2022

Open Access 01.12.2022 | Research

CUBN gene mutations may cause focal segmental glomerulosclerosis (FSGS) in children

verfasst von: Jing Yang, Yongli Xu, Linxia Deng, Luowen Zhou, Liru Qiu, Yu Zhang, Jianhua Zhou

Erschienen in: BMC Nephrology | Ausgabe 1/2022

Abstract

Background

Imerslund-Gräsbeck Syndrome (IGS) is mainly caused by CUBN gene biallelic mutations. Proteinuria accompanies IGS specific symptoms in about half of the patients, isolated proteinuria is rarely reported. Here we present 3 patients with isolated proteinuria and focal segmental glomerulosclerosis (FSGS) caused by CUBN gene biallelic pathogenic variants.

Method

Whole exome sequencing was performed on three children with isolated proteinuria. CUBN gene biallelic pathogenic variants were found and then verified by sanger sequencing. Their clinical, pathological and molecular genetic characteristics were analyzed and correlated accordingly.

Results

All three children presented with isolated proteinuria, no megaloblastic anemia. Their urine levels of β2 microglobulin were normal or slightly higher. Renal biopsies showed focal segmental glomerulosclerosis with mild glomerular mesangial hypercellularity, partial effacement of foot processes and podocyte microvillation. Two of them were found to carry compound heterozygous mutations and one homozygous mutation of CUBN gene. Totally four CUBN gene biallelic pathogenic variants were identified, including c.9287 T > C (p.L3096P), c.122 + 1G > A, c.7906C > T (p.R2636*), c.10233G > A (p.W3411*). Except for intron splice-site mutation, all other variants are located in highly conserved sites of CUB domain for binding to albumin.

Conclusion

The results demonstrate that CUBN gene mutations may cause isolated proteinuria pathologically presented as FSGS. Our cases extend the spectrum of renal manifestation and genotype of CUBN gene mutations.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
FSGS
Focal segmental glomerulosclerosis
CSA
Clinic Sequence Analyzer
ACMG
American College of Medical Genetics
IGS
Imerslund-Gräsbeck Syndrome
EM
Electron microscopy.

Introduction

Imerslund-Gräsbeck Syndrome (IGS) is a rare autosomal recessive disease caused by mutations of CUBN or AMN gene [1, 2]. The clinical features of IGS include selective intestinal vitamin B12 malabsorption resulting in megaloblastic anemia, failure to thrive, recurrent infections, neurological damage, with or without proteinuria and normal renal function [3] . To date, only 48 different mutations in CUBN and 32 different variants in AMN have been reported in the “Human Gene Mutation Database” (HGMD) (http://​www.​hgmd.​cf.​ac.​uk/​ac/​index.​php).
CUBN gene encodes a large 460-kDa glycosylated extracellular protein called cubilin, which is composed of 27 CUB domains [4]. Cubilin expresses in both renal proximal tubular cells [4] and podocytes [5]. It contains eight Ca2+-binding epidermal growth factor repeats and a N-terminal region involved in trimerization of the protein [6]. The CUB domains of cubulin can act as ligand-binding sites for intrinsic factor-B12 complex [7], albumin [8, 9], vitamin carrier proteins, lipoproteins, other carriers, immune- and stress-related proteins and drugs [10]. Among them, the intrinsic factor-B12 complex binds to CUB domains 5–8 [11], but the binding sites for albumin remain unclear. Cubilin has no transmembrane domain in the structure, so it is dependent on additional factors for membrane anchoring and for endocytosis of the receptor-bound ligands [6]. As an endocytic receptor, cubilin mediates the uptake of proteins and protein-bound substances both in intestine and kidney [6]. The majority of filtrated albumin is reabsorbed through cubilin-mediated endocytosis in the proximal tubule, resulting in very low level of albumin in final urine.
In addition to Imerslund-Gräsbeck Syndrome, CUBN gene mutations have been reported in only a few cases with isolated proteinuria. The proteinuria is thought from tubular loss without glomerular involvement and the prognosis is benign [1214]. In this paper, we report 3 patients with CUBN gene biallelic pathogenic variants presented as isolated proteinuria and FSGS in renal biopsy. Our cases extend the spectrum of renal manifestation and genotype of CUBN gene mutation.

Materials and methods

Patients

Patient 1 was a 11-year-old boy with asymptomatic persistent proteinuria of 2+ for more than one month with unknown causes. Patient 2 was a 6-year-old girl with foamy urine and proteinuria of 2+ for one year. Patient 3 was an 8-year-old boy with persistent proteinuria for more than one and half a year. All of them had no edema, hypertension, oliguria. Renal biopsy was performed and FSGS was reported in all of them.

Clinical data collection, renal histopathological evaluation and follow-up study

All three cases were regularly followed up and evaluated through examination of blood biochemistry, serum immunoglobulin and complement, urine albumin/creatinine and β2 microglobulin levels. Tacrolimus was tried in these cases after renal biopsy, started at dosage of 0.05–0.1 mg/kg and maintained a stable trough concentration at 5-10 ng/ml for at least 3 months.

Detection and analysis of CUBN gene variants by family trio whole exome sequencing

After the informed consent of the patients’ parents, we collected blood samples from the patients and their parents. Total DNA was extracted from peripheral blood using the QIAamp DNA Blood Kit (Qiagen) according to the kit standard protocol instruction. DNA were sheared with Covaris LE220 Sonicator (Covaris) to target of 150-200 bp average size. PCR amplification is performed using universal primers complementary to the adapter sequence to form a sequencing library. The sample genetic fingerprint was detected by Fluidigm Biomark using human specific SNP primers. The sequencing quality was determined by FastQC software performed on sequencing rawData. After data filtration, the clean reads were mapped to human reference genome (hg19) using Sentieon BWA software. Then, the mapped reads were detected to find SNV and InDel with Sentieon (the same algorithm with GATK) analysis, and annotated using ANNOVAR/VEP software. The pathogenic variants were screened by ClinVar, OMIM and HGMD. Functional prediction of missense mutation was determined by PolyPhen-2 and Sorting Intolerant from Tolerant (SIFT). Splice-cite prediction was determined by Human Splicing Finder (http: //www.​umd.​be/​HSF/​). The variants were evaluated by Clinic Sequence Analyzer (CSA) and classified according to “Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology” (published by ACMG, 2015), [15]. The variants were named according to the Human Genome Variation Society (HGVS) policy (https://​www.​hgvs.​org/​mutnomen). All the suspicious pathogenic variants were validated in patient and their parents by Sanger sequencing. The mutation sites were mapped in cubilin protein with DOG 2.0(Visualization of Protein Domain Structures) [16] and assessed for their conservation among different species using UCSC Genome Browser (http://​genome.​ucsc.​edu/​).
Sequences were submitted to the Swiss-Model server (https://​swissmodel.​expasy.​org/​) for modelling in automated mode, predicted effects of CUBN variants on protein structure were modelled using Swiss-PdbViewer [17].

Results

Clinical, renal pathological and follow-up findings

The clinical and renal pathological features were summarized in Table 1. All cases presented with proteinuria 2+ or over, urine β2 microglobulin was within the normal range in 2 cases and slightly increased in 1 case. Their urine protein electrophoresis showed albumin predominantly. Their hemoglobin (Hb) counts, serum IgG, IgA, IgM, C3, C4, and vitamin B12 levels were normal. All of them had normal renal functions, eGFR were 132, 165, 168 ml/min/1.73m2 respectively. Their renal ultrasonography showed no abnormalities. Renal biopsies showed segmental glomerulosclerosis (shown in Fig. 1 A), crescents (shown in Fig. 1 B, C), interstitial fibrosis (shown in Fig. 1 D). No immunoglobulin and complement deposited in the mesangial cells. Effacement of foot processes and podocyte microvillation were observed under electron microscopy (shown in Fig. 1 E, F). Neither of their parents has proteinuria, their family diagram of the three patients shown in Fig. 2. Urine albumin/creatinine values has decreased from 354μg/mg to 187μg/mg in patient 1, from 536μg/mg to 338μg/mg in patient 2, from 462μg/mg to 19.7μg/mg in patient 3 after administration of tacrolimus over three months.
Table 1
Clinical features of three patients
Patient
One
Two
Three
Age at onset (years)
10.7
5
6.5
Age at diagnosis (years)
11
6
8
Gender
Male
Female
Male
Edema
No
No
No
Megaloblastic anemia
No
No
No
proteinuria
2+
2+
2+
Urine protein/creatinine (ug/mg)
354
536
462
Urine β2-microglobulin
< 0.1 mg/L
< 0.1 mg/L
0.56 mg/L
Urinary protein electrophoresis
93.3% albumin
89.3% albumin
90% albumin
Hemoglobin(g/L)
115
116
130
Serum albumin
42.5
42.4
42.5
Egfr (ml/min/1.73m2)
132
165
168
LM
Mesangial cell proliferation
Mild
Mild
Mild
Segmental sclerosis (proportion)
2/24
1/40
1/55
Interstitial fibrosis
Yes
Yes
Yes
Crescent (number)
1
1
0
IF
No deposits
No deposits
No deposits
EM
Effacement of foot processes
Yes
Yes
Yes
Podocyte microvillation
Yes
Yes
Yes
Gene mutation
Allele 1
c.9287 T > C, p. L3096P (Exon59)
c.9287 T > C, p. L3096P(Exon59)
c.10233G > A, p.W3411*(Exon64)
Allele 2
c.122 + 1G > A (Intron1)
c.7906C > T, p.R2636* (Exon51)
c.10233G > A p.W3411*(Exon64)
eGFR Estimated glomerular filtration rate; LM Light microscopy; EM Electron microscopy; IF Immunofluorescence

Identification of CUBN gene mutation

CUBN gene biallelic pathogenic variants were identified by family trio WES (whole exome sequencing) of three children and their parents. Patient 1 carried compound heterozygous CUBN gene mutations c.9287 T > C (p.L3096P) and c.122 + 1G > A, which came from his father and mother respectively (shown in Fig. 3 A). The former was a missense mutation of CUBN gene, not seen in ExAC database (http://​exac.​broadinstitute.​org) and gnomAD database (https://​gnomad.​broadinstitute.​org). Poly-phen2, SIFT, Mutation Taster software revealed the scores were 0.813(possibly damaging), 0.003(damaging), and 1.000(disease causing) respectively. This missense mutation was regarded as possibly pathogenic (PM2, PM3, PP3, and PP4), the other splice-site mutation was predicted as “most probably affecting splicing” by Human Splicing Finder, and regarded as pathogenic (PVS1, PM2, and PP4) according to ACMG criteria.
Patient 2 carried compound heterozygous CUBN gene mutations c.9287 T > C (p.L3096P) and c.7906C > T (p.R2636*), which came from his father and mother respectively (shown in Fig. 3 B). The former was the same missense mutation as in patient 1. The other was a nonsense mutation leading to truncation of the protein. The frequency of this mutation was 0.00007 in ExAC and 9.339*10− 5 in gnomAD database. Mutation Taster, CADD and DANN software showed the mutation scores were 1.000(disease causing), 55(damaging), 0.998(damaging). The nonsense mutation was regarded as pathogenic (PVS1, PM2, and PP4) according to ACMG criteria.
Patient 3 carried homozygous CUBN gene mutations c.10233G > A (p. W3411*) (shown in Fig. 3 C). It was a nonsense mutation leading to cubulin truncation, not seen in ExAC but reported with frequency of 8.128*10− 6 in gnomAD database. Mutation Taster, CADD, DANN software revealed the mutation scores were 1.000(disease causing), 51(damaging), 0.993(damaging). This nonsense mutation was regarded as pathogenic (PVS1, PM2, and PP4) according to ACMG criteria.
Except for the splicing mutation in patient 1, all other mutations were located at CUB domains 19, 23, 27 of cubilin (shown in Fig. 3 D). UCSC Genome Browser alignment indicated that L3096, R2636 and W3411 in CUBN gene was highly conserved among different species (shown in Fig. 3 E). Since two patients shared the same missense mutation (c.9287 T > C, p.L3096P), we built 3D-structure model of the mutant cubilin protein based on the solved wild-type CUBN 3D-structure by Swiss-Model (shown in Fig. 4 A). They share high sequence identity (6fzv, 36.25%). The 3D-structure model and surface model between wild-type cubilin protein and mutant cubilin protein were showed in Fig. 4 B. The hydrogen bond was calculated and found to be changed in the mutant compared with wild-type.

Discussion

Previous studies have demonstrated that CUBN gene mutations can cause IGS that typically manifests as megaloblastic anemia and secondary neurological symptoms, with or without proteinuria. Isolated proteinuria caused by CUBN gene mutations is rare, only a few cases have been reported in the world so far. Boger et.al [18] found a missense mutation (I2984V) of CUBN gene which is associated with albuminuria in general population and in individuals with diabetes. In 2011, Ovunc et.al [3] identified a homozygous frameshift mutation in CUBN gene in two siblings of consanguineous patients with intermittent nephrotic-range proteinuria, which indicated that cubilin mutation may be considered as a rare single-gene cause of nephropathy. Recently Bedin et al. [14] identified 39 patients with biallelic CUBN gene mutations from three cohorts, and found that they had sub-nephrotic proteinuria and normal renal function. Our cases add more evidence that CUBN gene mutations may cause moderate proteinuria.
Cubilin works as a receptor for albumin, its defect may significantly reduce albumin reabsorption in renal proximal tubular cells and leads to the occurrence of proteinuria. Amsellem et.al [19] found that selective daily albumin excretion was increased approximately six-folds in cubilin-deficient mice. It is assumed that albuminuria come from renal tubular malabsorption in CUBN mutated patients, but whether it is also resulted from glomerular loss remains to be elucidated. Although there was no direct evidence to support glomerular origin, our cases showed pathological changes of podocytes and FSGS and their proteinuria gradually attenuated after administration of tacrolimus. Those suggest that albuminuria was probably not only originated from renal proximal tubule malabsorption, but may also from podocyte disfunction in CUBN-mutated patients.
Due to the scarcity of cases with CUBN gene mutations, kidney biopsy has been performed in only a few patients so far. Until now, focal segmental glomerulosclerosis has just been reported in 1 of 4 patients in genetic kidney disease cohort II with CUBN gene mutations [14]. In 2012, Prabakaran et.al [5] found cubilin expression in rat podocytes and human podocytes. Gianesello et.al [20] proved that cubilin mediates albumin endocytosis in human podocytes, CUBN gene mutations may lead to the dysfunction of cubilin, thus affect albumin endocytosis in podocytes. The development of proteinuria is usually related to podocyte damage, such as podocyte foot process effacement and cell loss [21]. Compared to megalin, cubilin is thought to have higher binding affinity for albumin [22]. Megalin can function as a sensor of albumin to determine the effect on cell survival. No or only a small amount of albumin binding to megalin can inhibit podocyte apoptosis, however, a large quantity of albumin binding to megalin can promote cell apoptosis [5]. It’s thought that the CUBN gene mutations lead to reduction of cubilin-binding ability with albumin, which make more free albumin available to bind with megalin. Consequently, it promotes podocyte apoptosis via the PI-3 K/PKB pathway, leading to a decreased number of podocytes. As a kind of terminal differentiated cell, podocyte can’t proliferate to compensate its loss, thereafter glomerular basement membrane become naked, followed by development of FSGS. Bedin et.al [14] reported the renal pathologies in 19 patients with CUBN gene mutations, most of them were minimal change disease or no lesions. But there were two patients whose lesions tended to be in the early stage of FSGS [14]. Interestingly, those pathologically manifested with FSGS unanimously had at least a relatively serious mutation in one allele, such as nonsense mutation, insertion, deletion or mutation in splice sites which led to frameshift or protein truncation. Since renal biopsy was rarely performed in patients with CUBN gene mutations, even less for electron microscopy (EM) examination. Therefore, the pathological changes of podocytes were not noted on EM in the past. The observation of obvious podocyte abnormalities with EM in our cases provide evidence for its involvement in the pathogenesis of albuminuria in CUBN gene-mutated patients.
Bedin et.al [14] firstly noted that all proteinuria-associated CUBN mutations were localized to C-terminal CUBN domains. Our patients showed that except for one intron splice-site mutation, other mutations are located in highly conserved sites at the C-terminal, CUB19, CUB23 and CUB27 domain respectively. All our cases had at least one relatively serious mutation and severe pathological changes accordingly.
In this study, four biallelic pathogenic variants of CUBN gene were identified in three isolated proteinuric children. The results demonstrate that CUBN gene mutations may cause isolated proteinuria pathologically presented as FSGS. Our cases extend the spectrum of renal manifestation and genotype of CUBN gene mutation.

Acknowledgements

We thank all patients and their families for the participation in this study.

Declarations

This study was approved by The Human Ethics Committees of Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology. Written informed assent was obtained from a parent or legal guardian for participants under 18 years old. All methods were performed in accordance with the relevant guidelines and regulations.
We obtained written informed consent for publication of the article, including any identifiable data and images from the parents of the three minors.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Aminoff M, et al. Mutations in CUBN, encoding the intrinsic factor-vitamin B12 receptor, cubilin, cause hereditary megaloblastic anaemia 1. Nat Genet. 1999;21:309–13.CrossRef Aminoff M, et al. Mutations in CUBN, encoding the intrinsic factor-vitamin B12 receptor, cubilin, cause hereditary megaloblastic anaemia 1. Nat Genet. 1999;21:309–13.CrossRef
2.
Zurück zum Zitat Tanner SM, et al. Amnionless, essential for mouse gastrulation, is mutated in recessive hereditary megaloblastic anemia. Nat Genet. 2003;33(3):426–9.CrossRef Tanner SM, et al. Amnionless, essential for mouse gastrulation, is mutated in recessive hereditary megaloblastic anemia. Nat Genet. 2003;33(3):426–9.CrossRef
3.
Zurück zum Zitat Ovunc B, et al. Exome sequencing reveals cubilin mutation as a single-gene cause of proteinuria. J Am Soc Nephrol. 2011;22(10):1815–20.CrossRef Ovunc B, et al. Exome sequencing reveals cubilin mutation as a single-gene cause of proteinuria. J Am Soc Nephrol. 2011;22(10):1815–20.CrossRef
4.
Zurück zum Zitat Nielsen R, Christensen EI, Birn H. Megalin and cubilin in proximal tubule protein reabsorption: from experimental models to human disease. Kidney Int. 2016;89(1):58–67.CrossRef Nielsen R, Christensen EI, Birn H. Megalin and cubilin in proximal tubule protein reabsorption: from experimental models to human disease. Kidney Int. 2016;89(1):58–67.CrossRef
5.
Zurück zum Zitat Prabakaran T, et al. Cubilin is expressed in rat and human glomerular podocytes. Nephrol Dial Transplant. 2012;27(8):3156–9.CrossRef Prabakaran T, et al. Cubilin is expressed in rat and human glomerular podocytes. Nephrol Dial Transplant. 2012;27(8):3156–9.CrossRef
6.
Zurück zum Zitat Christensen EI, Nielsen R, Birn H. From bowel to kidneys: the role of cubilin in physiology and disease. Nephrol Dial Transplant. 2013;28(2):274–81.CrossRef Christensen EI, Nielsen R, Birn H. From bowel to kidneys: the role of cubilin in physiology and disease. Nephrol Dial Transplant. 2013;28(2):274–81.CrossRef
7.
Zurück zum Zitat Christensen EI, Verroust PJ, Nielsen R. Receptor-mediated endocytosis in renal proximal tubule. Pflugers Arch. 2009;458(6):1039–48.CrossRef Christensen EI, Verroust PJ, Nielsen R. Receptor-mediated endocytosis in renal proximal tubule. Pflugers Arch. 2009;458(6):1039–48.CrossRef
8.
Zurück zum Zitat Birn H, et al. Cubilin is an albumin binding protein important for renal tubular albumin reabsorption. J Clin Invest. 2000;105:1353–61.CrossRef Birn H, et al. Cubilin is an albumin binding protein important for renal tubular albumin reabsorption. J Clin Invest. 2000;105:1353–61.CrossRef
9.
Zurück zum Zitat ZHAI XY, et al. Cubilin- and megalin-mediated uptake of albumin in cultured proximal tubule cells of opossum kidney. Kidney Int. 2000;2000(58):1523–33.CrossRef ZHAI XY, et al. Cubilin- and megalin-mediated uptake of albumin in cultured proximal tubule cells of opossum kidney. Kidney Int. 2000;2000(58):1523–33.CrossRef
10.
Zurück zum Zitat Nielsen R, Christensen EI. Proteinuria and events beyond the slit. Pediatr Nephrol. 2010;25(5):813–22.CrossRef Nielsen R, Christensen EI. Proteinuria and events beyond the slit. Pediatr Nephrol. 2010;25(5):813–22.CrossRef
11.
Zurück zum Zitat Andersen CB, et al. Structural basis for receptor recognition of vitamin-B (12)-intrinsic factor complexes. Nature. 2010;464(7287):445–8.CrossRef Andersen CB, et al. Structural basis for receptor recognition of vitamin-B (12)-intrinsic factor complexes. Nature. 2010;464(7287):445–8.CrossRef
12.
Zurück zum Zitat Ercan Z, et al. A long-term follow-up of an Imerslund-Grasbeck syndrome patient with proteinuria. Nefrologia. 2013;33(1):147–8.PubMed Ercan Z, et al. A long-term follow-up of an Imerslund-Grasbeck syndrome patient with proteinuria. Nefrologia. 2013;33(1):147–8.PubMed
13.
Zurück zum Zitat Grasbeck R. Imerslund-Grasbeck syndrome (selective vitamin B (12) malabsorption with proteinuria). Orphanet J Rare Dis. 2006;1:17.CrossRef Grasbeck R. Imerslund-Grasbeck syndrome (selective vitamin B (12) malabsorption with proteinuria). Orphanet J Rare Dis. 2006;1:17.CrossRef
14.
Zurück zum Zitat Bedin M, et al. Human C-terminal CUBN variants associate with chronic proteinuria and normal renal function. J Clin Invest. 2020;130(1):335–44.CrossRef Bedin M, et al. Human C-terminal CUBN variants associate with chronic proteinuria and normal renal function. J Clin Invest. 2020;130(1):335–44.CrossRef
15.
Zurück zum Zitat Richards S, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17(5):405–24.CrossRef Richards S, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17(5):405–24.CrossRef
16.
Zurück zum Zitat Ren J, et al. DOG 1.0: illustrator of protein domain structures. Cell Res. 2009;19(2):271–3.CrossRef Ren J, et al. DOG 1.0: illustrator of protein domain structures. Cell Res. 2009;19(2):271–3.CrossRef
17.
Zurück zum Zitat Guex N, Peitsch MC. SWISS-MODEL and the Swiss-PdbViewer:An environment for comparative protein modeling. Electrophoresis. 1997;18:2714–23.CrossRef Guex N, Peitsch MC. SWISS-MODEL and the Swiss-PdbViewer:An environment for comparative protein modeling. Electrophoresis. 1997;18:2714–23.CrossRef
18.
Zurück zum Zitat Boger CA, et al. CUBN is a gene locus for albuminuria. J Am Soc Nephrol. 2011;22(3):555–70.CrossRef Boger CA, et al. CUBN is a gene locus for albuminuria. J Am Soc Nephrol. 2011;22(3):555–70.CrossRef
19.
Zurück zum Zitat Amsellem S, et al. Cubilin is essential for albumin reabsorption in the renal proximal tubule. J Am Soc Nephrol. 2010;21(11):1859–67.CrossRef Amsellem S, et al. Cubilin is essential for albumin reabsorption in the renal proximal tubule. J Am Soc Nephrol. 2010;21(11):1859–67.CrossRef
20.
Zurück zum Zitat Gianesello L, et al. Albumin uptake in human podocytes: a possible role for the cubilin-amnionless (CUBAM) complex. Sci Rep. 2017;7(1):13705.CrossRef Gianesello L, et al. Albumin uptake in human podocytes: a possible role for the cubilin-amnionless (CUBAM) complex. Sci Rep. 2017;7(1):13705.CrossRef
21.
Zurück zum Zitat Liu Y, et al. Podocyte-released Migrasomes in urine serve as an Indicator for early podocyte injury. Kidney Dis (Basel). 2020;6(6):422–33.CrossRef Liu Y, et al. Podocyte-released Migrasomes in urine serve as an Indicator for early podocyte injury. Kidney Dis (Basel). 2020;6(6):422–33.CrossRef
22.
Zurück zum Zitat Christensen EI, et al. Endocytic receptors in the renal proximal tubule. Physiology (Bethesda). 2012;27(4):223–36. Christensen EI, et al. Endocytic receptors in the renal proximal tubule. Physiology (Bethesda). 2012;27(4):223–36.
Metadaten
Titel
CUBN gene mutations may cause focal segmental glomerulosclerosis (FSGS) in children
verfasst von
Jing Yang
Yongli Xu
Linxia Deng
Luowen Zhou
Liru Qiu
Yu Zhang
Jianhua Zhou
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
BMC Nephrology / Ausgabe 1/2022
Elektronische ISSN: 1471-2369
DOI
https://doi.org/10.1186/s12882-021-02654-x

Weitere Artikel der Ausgabe 1/2022

BMC Nephrology 1/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.