Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Clin Kidney J ; 17(8): sfae096, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39135942

ABSTRACT

Background: Idiopathic nephrotic syndrome (INS) is a heterogenous disease and current classification is based on observational responses to therapies or kidney histology. The National Unified Renal Translational Research Enterprise (NURTuRE)-INS cohort aims to facilitate novel ways of stratifying INS patients to improve disease understanding, therapeutics and design of clinical trials. Methods: NURTuRE-INS is a prospective cohort study of children and adults with INS in a linked biorepository. All recruits had at least one sampling visit collecting serum, plasma, urine and blood for RNA and DNA extraction, frozen within 2 hours of collection. Clinical histology slides and biopsy tissue blocks were also collected. Results: A total of 739 participants were recruited from 23 centres to NURTuRE-INS, half of whom were diagnosed in childhood [n = 365 (49%)]. The majority were white [n = 525 (71%)] and the median age at recruitment was 32 years (interquartile range 12-54). Steroid-sensitive nephrotic syndrome (SSNS) was the most common clinical diagnosis [n = 518 (70%)]. Of patients diagnosed in childhood who underwent a kidney biopsy, for SSNS (n =103), 76 demonstrated minimal change disease (MCD), whereas for steroid-resistant nephrotic syndrome (n =80), 21 had MCD. Almost all patients diagnosed in adulthood had a kidney biopsy [n = 352 (94%)]; 187 had MCD and 162 had focal segmental glomerulosclerosis. Conclusions: NURTuRE-INS is a prospective cohort study with high-quality biosamples and longitudinal data that will assist research into the mechanistic stratification of INS. Samples and data will be available through a Strategic Access and Oversight Committee.

2.
NPJ Syst Biol Appl ; 10(1): 28, 2024 Mar 09.
Article in English | MEDLINE | ID: mdl-38459044

ABSTRACT

Chronic kidney diseases (CKD) have genetic associations with kidney function. Univariate genome-wide association studies (GWAS) have identified single nucleotide polymorphisms (SNPs) associated with estimated glomerular filtration rate (eGFR) and blood urea nitrogen (BUN), two complementary kidney function markers. However, it is unknown whether additional SNPs for kidney function can be identified by multivariate statistical analysis. To address this, we applied canonical correlation analysis (CCA), a multivariate method, to two individual-level CKD genotype datasets, and metaCCA to two published GWAS summary statistics datasets. We identified SNPs previously associated with kidney function by published univariate GWASs with high replication rates, validating the metaCCA method. We then extended discovery and identified previously unreported lead SNPs for both kidney function markers, jointly. These showed expression quantitative trait loci (eQTL) colocalisation with genes having significant differential expression between CKD and healthy individuals. Several of these identified lead missense SNPs were predicted to have a functional impact, including in SLC14A2. We also identified previously unreported lead SNPs that showed significant correlation with both kidney function markers, jointly, in the European ancestry CKDGen, National Unified Renal Translational Research Enterprise (NURTuRE)-CKD and Salford Kidney Study (SKS) datasets. Of these, rs3094060 colocalised with FLOT1 gene expression and was significantly more common in CKD cases in both NURTURE-CKD and SKS, than in the general population. Overall, by using multivariate analysis by CCA, we identified additional SNPs and genes for both kidney function and CKD, that can be prioritised for further CKD analyses.


Subject(s)
Genome-Wide Association Study , Renal Insufficiency, Chronic , Humans , Genome-Wide Association Study/methods , Canonical Correlation Analysis , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/epidemiology , Kidney , Quantitative Trait Loci/genetics
4.
Pediatr Nephrol ; 38(6): 1793-1800, 2023 06.
Article in English | MEDLINE | ID: mdl-36357634

ABSTRACT

BACKGROUND: Idiop athic nephrotic syndrome (INS) is classified in children according to response to initial corticosteroid therapy into steroid-sensitive (SSNS) and steroid-resistant nephrotic syndrome (SRNS), and in adults according to histology into minimal change disease (MCD) and focal segmental glomerulosclerosis (FSGS). However, there is well-recognised phenotypic overlap between these entities. Genome-wide association studies (GWAS) have shown a strong association between SSNS and variation at HLA, suggesting an underlying immunological basis. We sought to determine whether a risk score generated from genetic variants associated with SSNS could be used to gain insight into the pathophysiology of INS presenting in other ways. METHODS: We developed an SSNS genetic risk score (SSNS-GRS) from the five variants independently associated with childhood SSNS in a previous European GWAS. We quantified SSNS-GRS in independent cohorts of European individuals with childhood SSNS, non-monogenic SRNS, MCD, and FSGS, and contrasted them with SSNS-GRS quantified in individuals with monogenic SRNS, membranous nephropathy (a different immune-mediated disease-causing nephrotic syndrome), and healthy controls. RESULTS: The SSNS-GRS was significantly elevated in cohorts with SSNS, non-monogenic SRNS, MCD, and FSGS compared to healthy participants and those with membranous nephropathy. The SSNS-GRS in all cohorts with non-monogenic INS were also significantly elevated compared to those with monogenic SRNS. CONCLUSIONS: The shared genetic risk factors among patients with different presentations of INS strongly suggests a shared autoimmune pathogenesis when monogenic causes are excluded. Use of the SSNS-GRS, in addition to testing for monogenic causes, may help to classify patients presenting with INS. A higher resolution version of the Graphical abstract is available as Supplementary information.


Subject(s)
Glomerulonephritis, Membranous , Glomerulosclerosis, Focal Segmental , Nephrosis, Lipoid , Nephrotic Syndrome , Child , Humans , Nephrotic Syndrome/diagnosis , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/genetics , Nephrosis, Lipoid/diagnosis , Nephrosis, Lipoid/drug therapy , Nephrosis, Lipoid/genetics , Glomerulosclerosis, Focal Segmental/diagnosis , Glomerulosclerosis, Focal Segmental/drug therapy , Glomerulosclerosis, Focal Segmental/genetics , Genome-Wide Association Study , Steroids , Risk Factors
5.
Pediatr Nephrol ; 37(11): 2643-2656, 2022 11.
Article in English | MEDLINE | ID: mdl-35211795

ABSTRACT

BACKGROUND: Variants in genes encoding nuclear pore complex (NPC) proteins are a newly identified cause of paediatric steroid-resistant nephrotic syndrome (SRNS). Recent reports describing NUP93 variants suggest these could be a significant cause of paediatric onset SRNS. We report NUP93 cases in the UK and demonstrate in vivo functional effects of Nup93 depletion in a fly (Drosophila melanogaster) nephrocyte model. METHODS: Three hundred thirty-seven paediatric SRNS patients from the National cohort of patients with Nephrotic Syndrome (NephroS) were whole exome and/or whole genome sequenced. Patients were screened for over 70 genes known to be associated with Nephrotic Syndrome (NS). D. melanogaster Nup93 knockdown was achieved by RNA interference using nephrocyte-restricted drivers. RESULTS: Six novel homozygous and compound heterozygous NUP93 variants were detected in 3 sporadic and 2 familial paediatric onset SRNS characterised histologically by focal segmental glomerulosclerosis (FSGS) and progressing to kidney failure by 12 months from clinical diagnosis. Silencing of the two orthologs of human NUP93 expressed in D. melanogaster, Nup93-1, and Nup93-2 resulted in significant signal reduction of up to 82% in adult pericardial nephrocytes with concomitant disruption of NPC protein expression. Additionally, nephrocyte morphology was highly abnormal in Nup93-1 and Nup93-2 silenced flies surviving to adulthood. CONCLUSION: We expand the spectrum of NUP93 variants detected in paediatric onset SRNS and demonstrate its incidence within a national cohort. Silencing of either D. melanogaster Nup93 ortholog caused a severe nephrocyte phenotype, signaling an important role for the nucleoporin complex in podocyte biology. A higher resolution version of the Graphical abstract is available as Supplementary information.


Subject(s)
Drosophila melanogaster , Nephrotic Syndrome , Nuclear Pore Complex Proteins , Podocytes , Adult , Animals , Child , Disease Models, Animal , Drosophila melanogaster/genetics , Drug Resistance/genetics , Glucocorticoids/adverse effects , Glucocorticoids/therapeutic use , High-Throughput Nucleotide Sequencing , Humans , Mutation , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/genetics , Nephrotic Syndrome/metabolism , Nuclear Pore Complex Proteins/genetics , Podocytes/metabolism
6.
Clin J Am Soc Nephrol ; 17(1): 143-154, 2022 01.
Article in English | MEDLINE | ID: mdl-34930753

ABSTRACT

Genetic testing for pathogenic COL4A3-5 variants is usually undertaken to investigate the cause of persistent hematuria, especially with a family history of hematuria or kidney function impairment. Alport syndrome experts now advocate genetic testing for persistent hematuria, even when a heterozygous pathogenic COL4A3 or COL4A4 is suspected, and cascade testing of their first-degree family members because of their risk of impaired kidney function. The experts recommend too that COL4A3 or COL4A4 heterozygotes do not act as kidney donors. Testing for variants in the COL4A3-COL4A5 genes should also be performed for persistent proteinuria and steroid-resistant nephrotic syndrome due to suspected inherited FSGS and for familial IgA glomerulonephritis and kidney failure of unknown cause.


Subject(s)
Autoantigens/genetics , Collagen Type IV/genetics , Genetic Testing/standards , Nephritis, Hereditary/diagnosis , Nephritis, Hereditary/genetics , Nephritis, Hereditary/therapy , Humans , Practice Guidelines as Topic
7.
Endocrinology ; 163(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-34791124

ABSTRACT

Diabetic nephropathy (DN) is the leading cause of kidney failure, with an increasing incidence worldwide. Mitochondrial dysfunction is known to occur in DN and has been implicated in the underlying pathogenesis of disease. These complex organelles have an array of important cellular functions and involvement in signaling pathways, and understanding the intricacies of these responses in health, as well as how they are damaged in disease, is likely to highlight novel therapeutic avenues. A key cell type damaged early in DN is the podocyte, and increasing studies have focused on investigating the role of mitochondria in podocyte injury. This review will summarize what is known about podocyte mitochondrial dynamics in DN, with a particular focus on bioenergetic pathways, highlighting key studies in this field and potential opportunities to target, enhance or protect podocyte mitochondrial function in the treatment of DN.


Subject(s)
Diabetic Nephropathies/metabolism , Energy Metabolism , Mitochondria/metabolism , Podocytes/metabolism , Animals , Diabetes Mellitus, Experimental/metabolism , Humans , Mitochondrial Dynamics , Mitophagy , Signal Transduction , Stress, Physiological
8.
Pediatr Nephrol ; 36(11): 3757-3769, 2021 11.
Article in English | MEDLINE | ID: mdl-34031708

ABSTRACT

BACKGROUND: Genetic defects in podocyte proteins account for up to 30% of steroid-resistant nephrotic syndrome (SRNS) in the paediatric population. Most children with genetic SRNS are resistant to immunosuppression and at high risk of progression to stage 5 chronic kidney disease. Kidney transplantation is often the treatment of choice. The possibility of post-transplantation disease recurrence in genetic SRNS remains controversial, and poses fundamental questions about disease biology. METHODS: We critically evaluated the published cases of post-transplantation recurrence in genetic patients, particularly testing 'mutations' against the most recent population variant databases, in order to clarify the diagnoses, and compare the clinical courses and responses to therapy. RESULTS: Biallelic pathogenic variants in NPHS1 leading to a complete absence of nephrin were the most commonly reported and best understood instance of nephrotic syndrome occurring post-transplantation. This is an immune-mediated process driven by antibody production against the novel nephrin protein in the allograft. We also identified a number of plausible reported cases of post-transplantation recurrence involving pathogenic variants in NPHS2 (8 patients, biallelic), one in WT1 (monoallelic) and one in NUP93 (biallelic). However, the mechanism for recurrence in these cases remains unclear. Other instances of recurrence in genetic disease were difficult to interpret due to differing clinical criteria, inclusion of patients without true pathogenic variants or the influence of other factors on renal outcome. CONCLUSIONS: Overall, post-transplantation recurrence remains very rare in patients with genetic SRNS. It appears to occur later after transplantation than in other patients and usually responds well to plasmapheresis with a good renal outcome.


Subject(s)
Kidney Transplantation , Nephrotic Syndrome , Child , Humans , Kidney Transplantation/adverse effects , Nephrotic Syndrome/genetics , Nephrotic Syndrome/pathology , Recurrence
9.
Eur J Hum Genet ; 29(8): 1186-1197, 2021 08.
Article in English | MEDLINE | ID: mdl-33854215

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.


Subject(s)
Consensus , Genetic Testing/methods , Nephritis, Hereditary/genetics , Practice Guidelines as Topic , Autoantigens/genetics , Collagen Type IV/genetics , Genetic Testing/standards , Humans , Nephritis, Hereditary/diagnosis , Phenotype
10.
J Am Soc Nephrol ; 32(7): 1682-1695, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33863784

ABSTRACT

BACKGROUND: Podocyte dysfunction is the main pathologic mechanism driving the development of FSGS and other morphologic types of steroid-resistant nephrotic syndrome (SRNS). Despite significant progress, the genetic causes of most cases of SRNS have yet to be identified. METHODS: Whole-genome sequencing was performed on 320 individuals from 201 families with familial and sporadic NS/FSGS with no pathogenic mutations in any known NS/FSGS genes. RESULTS: Two variants in the gene encoding regulator of calcineurin type 1 (RCAN1) segregate with disease in two families with autosomal dominant FSGS/SRNS. In vitro, loss of RCAN1 reduced human podocyte viability due to increased calcineurin activity. Cells expressing mutant RCAN1 displayed increased calcineurin activity and NFAT activation that resulted in increased susceptibility to apoptosis compared with wild-type RCAN1. Treatment with GSK-3 inhibitors ameliorated this elevated calcineurin activity, suggesting the mutation alters the balance of RCAN1 regulation by GSK-3ß, resulting in dysregulated calcineurin activity and apoptosis. CONCLUSIONS: These data suggest mutations in RCAN1 can cause autosomal dominant FSGS. Despite the widespread use of calcineurin inhibitors in the treatment of NS, genetic mutations in a direct regulator of calcineurin have not been implicated in the etiology of NS/FSGS before this report. The findings highlight the therapeutic potential of targeting RCAN1 regulatory molecules, such as GSK-3ß, in the treatment of FSGS.

11.
Am J Hum Genet ; 108(2): 357-367, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33508234

ABSTRACT

Focal segmental glomerulosclerosis (FSGS) is the main pathology underlying steroid-resistant nephrotic syndrome (SRNS) and a leading cause of chronic kidney disease. Monogenic forms of pediatric SRNS are predominantly caused by recessive mutations, while the contribution of de novo variants (DNVs) to this trait is poorly understood. Using exome sequencing (ES) in a proband with FSGS/SRNS, developmental delay, and epilepsy, we discovered a nonsense DNV in TRIM8, which encodes the E3 ubiquitin ligase tripartite motif containing 8. To establish whether TRIM8 variants represent a cause of FSGS, we aggregated exome/genome-sequencing data for 2,501 pediatric FSGS/SRNS-affected individuals and 48,556 control subjects, detecting eight heterozygous TRIM8 truncating variants in affected subjects but none in control subjects (p = 3.28 × 10-11). In all six cases with available parental DNA, we demonstrated de novo inheritance (p = 2.21 × 10-15). Reverse phenotyping revealed neurodevelopmental disease in all eight families. We next analyzed ES from 9,067 individuals with epilepsy, yielding three additional families with truncating TRIM8 variants. Clinical review revealed FSGS in all. All TRIM8 variants cause protein truncation clustering within the last exon between residues 390 and 487 of the 551 amino acid protein, indicating a correlation between this syndrome and loss of the TRIM8 C-terminal region. Wild-type TRIM8 overexpressed in immortalized human podocytes and neuronal cells localized to nuclear bodies, while constructs harboring patient-specific variants mislocalized diffusely to the nucleoplasm. Co-localization studies demonstrated that Gemini and Cajal bodies frequently abut a TRIM8 nuclear body. Truncating TRIM8 DNVs cause a neuro-renal syndrome via aberrant TRIM8 localization, implicating nuclear bodies in FSGS and developmental brain disease.


Subject(s)
Carrier Proteins/genetics , Developmental Disabilities/genetics , Epilepsy/genetics , Glomerulosclerosis, Focal Segmental/genetics , Intranuclear Space/metabolism , Nephrotic Syndrome/genetics , Nephrotic Syndrome/metabolism , Nerve Tissue Proteins/genetics , Adult , Animals , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Line , Child , Child, Preschool , Codon, Nonsense , Developmental Disabilities/metabolism , Epilepsy/metabolism , Female , Glomerulosclerosis, Focal Segmental/metabolism , Humans , Kidney/metabolism , Male , Mice , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Phenotype , Podocytes/metabolism , Exome Sequencing
13.
Clin J Am Soc Nephrol ; 15(7): 983-994, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32317330

ABSTRACT

BACKGROUND AND OBJECTIVES: Intensified immunosuppression in steroid-resistant nephrotic syndrome is broadly applied, with disparate outcomes. This review of patients from the United Kingdom National Study of Nephrotic Syndrome cohort aimed to improve disease stratification by determining, in comprehensively genetically screened patients with steroid-resistant nephrotic syndrome, if there is an association between response to initial intensified immunosuppression and disease progression and/or post-transplant recurrence. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Pediatric patients with steroid-resistant nephrotic syndrome were recruited via the UK National Registry of Rare Kidney Diseases. All patients were whole-genome sequenced, whole-exome sequenced, or steroid-resistant nephrotic syndrome gene-panel sequenced. Complete response or partial response within 6 months of starting intensified immunosuppression was ascertained using laboratory data. Response to intensified immunosuppression and outcomes were analyzed according to genetic testing results, pattern of steroid resistance, and first biopsy findings. RESULTS: Of 271 patients, 178 (92 males, median onset age 4.7 years) received intensified immunosuppression with response available. A total of 4% of patients with monogenic disease showed complete response, compared with 25% of genetic-testing-negative patients (P=0.02). None of the former recurred post-transplantation. In genetic-testing-negative patients, 97% with complete response to first intensified immunosuppression did not progress, whereas 44% of nonresponders developed kidney failure with 73% recurrence post-transplant. Secondary steroid resistance had a higher complete response rate than primary/presumed resistance (43% versus 23%; P=0.001). The highest complete response rate in secondary steroid resistance was to rituximab (64%). Biopsy results showed no correlation with intensified immunosuppression response or outcome. CONCLUSIONS: Patients with monogenic steroid-resistant nephrotic syndrome had a poor therapeutic response and no post-transplant recurrence. In genetic-testing-negative patients, there was an association between response to first intensified immunosuppression and long-term outcome. Patients with complete response rarely progressed to kidney failure, whereas nonresponders had poor kidney survival and a high post-transplant recurrence rate. Patients with secondary steroid resistance were more likely to respond, particularly to rituximab.


Subject(s)
Immunosuppressive Agents/therapeutic use , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/genetics , Rituximab/therapeutic use , Adolescent , Child , Child, Preschool , Cyclophosphamide/therapeutic use , Cyclosporine/therapeutic use , Disease Progression , Drug Resistance , Female , Genetic Testing , Humans , Immunosuppression Therapy/methods , Infant , Infant, Newborn , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/surgery , Kidney Transplantation , Male , Nephrotic Syndrome/pathology , Nephrotic Syndrome/surgery , Postoperative Period , Recurrence , Steroids , Tacrolimus/therapeutic use , Treatment Outcome
14.
Pediatr Nephrol ; 35(4): 641-648, 2020 04.
Article in English | MEDLINE | ID: mdl-31811534

ABSTRACT

BACKGROUND: Lowe syndrome and Dent-2 disease are caused by mutations in the OCRL gene, which encodes for an inositol 5-phosphatase. The renal phenotype associated with OCRL mutations typically comprises a selective proximal tubulopathy, which can manifest as Fanconi syndrome in the most extreme cases. METHODS: Here, we report a 12-year-old male with nephrotic-range proteinuria and focal segmental glomerulosclerosis on renal biopsy. As a glomerular pathology was suspected, extensive investigation of tubular function was not performed. RESULTS: Surprisingly, whole exome sequencing identified a genetic variant in OCRL (c1467-2A>G) that introduced a novel splice mutation leading to skipping of exon 15. In situ hybridisation of adult human kidney tissue and zebrafish larvae showed OCRL expression in the glomerulus, supporting a role for OCRL in glomerular function. In cultured podocytes, we found that OCRL associated with the linker protein IPIP27A and CD2AP, a protein that is important for maintenance of the podocyte slit diaphragm. CONCLUSION: Taken together, this work suggests a previously under-appreciated role for OCRL in glomerular function and highlights the importance of investigating tubular function in patients with persistent proteinuria.


Subject(s)
Glomerulosclerosis, Focal Segmental/genetics , Kidney Glomerulus/metabolism , Oculocerebrorenal Syndrome/genetics , Animals , Child , Chloride Channels , Glomerulosclerosis, Focal Segmental/complications , Humans , Male , Mutation , Oculocerebrorenal Syndrome/complications , Phosphoric Monoester Hydrolases , Podocytes/metabolism , Proteinuria/etiology , Exome Sequencing , Zebrafish
15.
Kidney Int ; 96(3): 642-655, 2019 09.
Article in English | MEDLINE | ID: mdl-31171376

ABSTRACT

The essential role of membrane associated guanylate kinase 2 (MAGI2) in podocytes is indicated by the phenotypes of severe glomerulosclerosis of both MAGI2 knockout mice and in patients with congenital nephrotic syndrome (CNS) caused by mutations in MAGI2. Here, we show that MAGI2 forms a complex with the Rap1 guanine nucleotide exchange factor, RapGEF2, and that this complex is lost when expressing MAGI2 CNS variants. Co-expression of RapGEF2 with wild-type MAGI2, but not MAGI2 CNS variants, enhanced activation of the small GTPase Rap1, a central signaling node in podocytes. In mice, podocyte-specific RapGEF2 deletion resulted in spontaneous glomerulosclerosis, with qualitative glomerular features comparable to MAGI2 knockout mice. Knockdown of RapGEF2 or MAGI2 in human podocytes caused similar reductions in levels of Rap1 activation and Rap1-mediated downstream signaling. Furthermore, human podocytes expressing MAGI2 CNS variants show severe abnormalities of cellular morphology and dramatic loss of actin cytoskeletal organization, features completely rescued by pharmacological activation of Rap1 via a non-MAGI2 dependent upstream pathway. Finally, immunostaining of kidney sections from patients with congenital nephrotic syndrome and MAGI2 mutations showed reduced podocyte Rap1-mediated signaling. Thus, MAGI2-RapGEF2-Rap1 signaling is essential for normal podocyte function. Hence, disruption of this pathway is an important cause of the renal phenotype induced by MAGI2 CNS mutations.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Guanine Nucleotide Exchange Factors/metabolism , Guanylate Kinases/genetics , Nephrotic Syndrome/genetics , Nerve Tissue Proteins/metabolism , Podocytes/pathology , Telomere-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Guanine Nucleotide Exchange Factors/genetics , Guanylate Kinases/metabolism , Humans , Mice , Mice, Knockout , Mutation , Nephrotic Syndrome/pathology , Shelterin Complex , Signal Transduction/drug effects , Signal Transduction/genetics , Telomere-Binding Proteins/agonists , rap1 GTP-Binding Proteins/metabolism
16.
Am J Hum Genet ; 104(2): 348-355, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30661770

ABSTRACT

Steroid-resistant nephrotic syndrome (SRNS) is characterized by high-range proteinuria and most often focal and segmental glomerulosclerosis (FSGS). Identification of mutations in genes causing SRNS has improved our understanding of disease mechanisms and highlighted defects in the podocyte, a highly specialized glomerular epithelial cell, as major factors in disease pathogenesis. By exome sequencing, we identified missense mutations in TBC1D8B in two families with an X-linked early-onset SRNS with FSGS. TBC1D8B is an uncharacterized Rab-GTPase-activating protein likely involved in endocytic and recycling pathways. Immunofluorescence studies revealed TBC1D8B presence in human glomeruli, and affected individual podocytes displayed architectural changes associated with migration defects commonly found in FSGS. In zebrafish we demonstrated that both knockdown and knockout of the unique TBC1D8B ortholog-induced proteinuria and that this phenotype was rescued by human TBC1D8B mRNA injection, but not by either of the two mutated mRNAs. We also showed an interaction between TBC1D8B and Rab11b, a key protein in vesicular recycling in cells. Interestingly, both internalization and recycling processes were dramatically decreased in affected individuals' podocytes and fibroblasts, confirming the crucial role of TBC1D8B in the cellular recycling processes, probably as a Rab11b GTPase-activating protein. Altogether, these results confirmed that pathogenic variations in TBC1D8B are involved in X-linked podocytopathy and points to alterations in recycling processes as a mechanism of SRNS.


Subject(s)
Calcium-Binding Proteins/genetics , Genetic Diseases, X-Linked/genetics , Loss of Function Mutation , Nephrotic Syndrome/genetics , Vesicular Transport Proteins/genetics , Zebrafish Proteins/genetics , Animals , Biological Transport/genetics , Calcium-Binding Proteins/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Kidney Glomerulus/metabolism , Male , Podocytes/cytology , Podocytes/metabolism , Vesicular Transport Proteins/metabolism , Exome Sequencing , Zebrafish , Zebrafish Proteins/metabolism , rab GTP-Binding Proteins/metabolism
17.
Pediatr Nephrol ; 33(11): 2027-2035, 2018 11.
Article in English | MEDLINE | ID: mdl-29022104

ABSTRACT

After renal transplantation, recurrence of the original disease is the second most common cause of graft loss, after rejection. The most dramatic manifestation of this phenomenon is in patients with nephrotic syndrome (NS). NS is a descriptive term describing a clinical picture centred on proteinuria arising from damage to the glomerular filtration barrier (GFB). There are many different drivers of that damage, ranging from immune dysregulation to genetic disorders and chronic disease/infections. The main categories in childhood are "idiopathic" (presumed immune mediated) and genetic NS, with further stratification of the idiopathic group according to steroid responses. A significant proportion of patients with NS progress to established renal failure, requiring transplantation, and one of the most difficult clinical scenarios faced by nephrologists is the recurrence of the original disease in up to 50% of patients, usually rapidly post-transplant. This is thought to be the archetypal "circulating factor" disease, in which as yet unknown circulating plasma "factor(s)" in the recipient target the donor kidney. The ability to predict in advance which patients will suffer recurrence would enhance our ability to counsel patients and families, and potentially identify those patients before transplant for tailored immunosuppressive preparation. Until very recently, stratification based on clinical categorisations has been poor in being able to predict those patients in whom disease will recur, and laboratory biomarkers are yet to be adequately refined. However, by mapping our growing understanding of disease mechanisms to clinical phenotypes, and with greatly improved genetic diagnostics, we have made progress in being able to stratify patients more specifically, and allow better predictive algorithms to be developed. Using our knowledge of podocyte biology, circulating factor-induced specific biomarkers are also being tested. This review is aimed at outlining those advances, and suggesting how we can move further forward in both clinical and biological markers of disease type.


Subject(s)
Allografts/pathology , Kidney Transplantation/adverse effects , Nephrotic Syndrome/diagnosis , Podocytes/pathology , Recurrence , Allografts/cytology , Biomarkers/analysis , Genetic Testing , Graft Survival , Humans , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/surgery , Mutation , Nephrotic Syndrome/complications , Nephrotic Syndrome/genetics , Nephrotic Syndrome/pathology , Risk Assessment/methods
18.
J Med Genet ; 54(12): 795-804, 2017 12.
Article in English | MEDLINE | ID: mdl-28780565

ABSTRACT

BACKGROUND: There are many single-gene causes of steroid-resistant nephrotic syndrome (SRNS) and the list continues to grow rapidly. Prompt comprehensive diagnostic testing is key to realising the clinical benefits of a genetic diagnosis. This report describes a bespoke-designed, targeted next-generation sequencing (NGS) diagnostic gene panel assay to detect variants in 37 genes including the ability to identify copy number variants (CNVs). METHODS: This study reports results of 302 patients referred for SRNS diagnostic gene panel analysis. Phenotype and clinical impact data were collected using a standard proforma. Candidate variants detected by NGS were confirmed by Sanger sequencing/Multiplex Ligation-dependent Probe Amplification with subsequent family segregation analysis where possible. RESULTS: Clinical presentation was nephrotic syndrome in 267 patients and suspected Alport syndrome (AS) in 35. NGS panel testing determined a likely genetic cause of disease in 44/220 (20.0%) paediatric and 10/47 (21.3%) adult nephrotic cases, and 17/35 (48.6%) of haematuria/AS patients. Of 71 patients with genetic disease, 32 had novel pathogenic variants without a previous disease association including two with deletions of one or more exons of NPHS1 or NPHS2. CONCLUSION: Gene panel testing provides a genetic diagnosis in a significant number of patients presenting with SRNS or suspected AS. It should be undertaken at an early stage of the care pathway and include the ability to detect CNVs as an emerging mechanism for genes associated with this condition. Use of clinical genetic testing after diagnosis of SRNS has the potential to stratify patients and assist decision-making regarding management.


Subject(s)
Drug Resistance/genetics , Genetic Testing , Nephrotic Syndrome/diagnosis , Nephrotic Syndrome/genetics , Adolescent , Age of Onset , Child , Child, Preschool , Female , Gene Expression Profiling/methods , Genetic Association Studies , Genetic Testing/methods , Genetic Variation , Humans , Infant , Infant, Newborn , Male , Nephrotic Syndrome/drug therapy , Phenotype , Steroids/therapeutic use , Young Adult
19.
F1000Res ; 6: 121, 2017.
Article in English | MEDLINE | ID: mdl-28232870

ABSTRACT

Idiopathic nephrotic syndrome (INS) is one of the most common glomerular diseases in children and adults, and the central event is podocyte injury. INS is a heterogeneous disease, and treatment is largely empirical and in many cases unsuccessful, and steroids are the initial mainstay of therapy. Close to 70% of children with INS have some response to steroids and are labelled as steroid-'sensitive', and the rest as steroid-'resistant' (also termed focal segmental glomerulosclerosis), and single-gene mutations underlie a large proportion of the latter group. The burden of morbidity is enormous, both to patients with lifelong chronic disease and to health services, particularly in managing dialysis and transplantation. The target cell of nephrotic syndrome is the glomerular podocyte, and podocyte biology research has exploded over the last 15 years. Major advances in genetic and biological understanding now put clinicians and researchers at the threshold of a major reclassification of the disease and testing of targeted therapies both identified and novel. That potential is based on complete genetic analysis, deep clinical phenotyping, and the introduction of mechanism-derived biomarkers into clinical practice. INS can now be split off into those with a single-gene defect, of which currently at least 53 genes are known to be causative, and the others. Of the others, the majority are likely to be immune-mediated and caused by the presence of a still-unknown circulating factor or factors, and whether there is a third (or more) mechanistic group or groups remains to be discovered. Treatment is therefore now being refined towards separating out the monogenic cases to minimise immunosuppression and further understanding how best to stratify and appropriately direct immunosuppressive treatments within the immune group. Therapies directed specifically towards the target cell, the podocyte, are in their infancy but hold considerable promise for the near future.

20.
J Clin Invest ; 127(3): 942-953, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28165343

ABSTRACT

Primary adrenal insufficiency is life threatening and can present alone or in combination with other comorbidities. Here, we have described a primary adrenal insufficiency syndrome and steroid-resistant nephrotic syndrome caused by loss-of-function mutations in sphingosine-1-phosphate lyase (SGPL1). SGPL1 executes the final decisive step of the sphingolipid breakdown pathway, mediating the irreversible cleavage of the lipid-signaling molecule sphingosine-1-phosphate (S1P). Mutations in other upstream components of the pathway lead to harmful accumulation of lysosomal sphingolipid species, which are associated with a series of conditions known as the sphingolipidoses. In this work, we have identified 4 different homozygous mutations, c.665G>A (p.R222Q), c.1633_1635delTTC (p.F545del), c.261+1G>A (p.S65Rfs*6), and c.7dupA (p.S3Kfs*11), in 5 families with the condition. In total, 8 patients were investigated, some of whom also manifested other features, including ichthyosis, primary hypothyroidism, neurological symptoms, and cryptorchidism. Sgpl1-/- mice recapitulated the main characteristics of the human disease with abnormal adrenal and renal morphology. Sgpl1-/- mice displayed disrupted adrenocortical zonation and defective expression of steroidogenic enzymes as well as renal histology in keeping with a glomerular phenotype. In summary, we have identified SGPL1 mutations in humans that perhaps represent a distinct multisystemic disorder of sphingolipid metabolism.


Subject(s)
Adrenal Insufficiency/congenital , Aldehyde-Lyases/genetics , Homozygote , INDEL Mutation , Mutation, Missense , Nephrotic Syndrome/genetics , Adrenal Glands/enzymology , Adrenal Glands/pathology , Adrenal Insufficiency/enzymology , Adrenal Insufficiency/genetics , Adrenal Insufficiency/pathology , Aldehyde-Lyases/metabolism , Animals , HEK293 Cells , Humans , Kidney/enzymology , Kidney/pathology , Mice , Mice, Knockout , Nephrotic Syndrome/enzymology , Nephrotic Syndrome/pathology
SELECTION OF CITATIONS
SEARCH DETAIL