Skip to main content
Erschienen in: Orphanet Journal of Rare Diseases 1/2016

Open Access 01.12.2016 | Research

Increased Wnt and Notch signaling: a clue to the renal disease in Schimke immuno-osseous dysplasia?

verfasst von: Marie Morimoto, Clara Myung, Kimberly Beirnes, Kunho Choi, Yumi Asakura, Arend Bokenkamp, Dominique Bonneau, Milena Brugnara, Joel Charrow, Estelle Colin, Amira Davis, Georges Deschenes, Mattia Gentile, Mario Giordano, Andrew K. Gormley, Rajeshree Govender, Mark Joseph, Kory Keller, Evelyne Lerut, Elena Levtchenko, Laura Massella, Christy Mayfield, Behzad Najafian, David Parham, Jurgen Spranger, Peter Stenzel, Uluc Yis, Zhongxin Yu, Jonathan Zonana, Glenda Hendson, Cornelius F. Boerkoel

Erschienen in: Orphanet Journal of Rare Diseases | Ausgabe 1/2016

Abstract

Background

Schimke immuno-osseous dysplasia (SIOD) is a multisystemic disorder caused by biallelic mutations in the SWI/SNF-related matrix-associated actin-dependent regulator of chromatin, subfamily A-like 1 (SMARCAL1) gene. Changes in gene expression underlie the arteriosclerosis and T-cell immunodeficiency of SIOD; therefore, we hypothesized that SMARCAL1 deficiency causes the focal segmental glomerulosclerosis (FSGS) of SIOD by altering renal gene expression. We tested this hypothesis by gene expression analysis of an SIOD patient kidney and verified these findings through immunofluorescent analysis in additional SIOD patients and a genetic interaction analysis in Drosophila.

Results

We found increased expression of components and targets of the Wnt and Notch signaling pathways in the SIOD patient kidney, increased levels of unphosphorylated β-catenin and Notch1 intracellular domain in the glomeruli of most SIOD patient kidneys, and genetic interaction between the Drosophila SMARCAL1 homologue Marcal1 and genes of the Wnt and Notch signaling pathways.

Conclusions

We conclude that increased Wnt and Notch activity result from SMARCAL1 deficiency and, as established causes of FSGS, contribute to the renal disease of most SIOD patients. This further clarifies the pathogenesis of SIOD and will hopefully direct potential therapeutic approaches for SIOD patients.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s13023-016-0519-7) contains supplementary material, which is available to authorized users.
Abkürzungen
ACV
Anterior crossvein
DAPI
4', 6-diamidino-2-phenylindole
DAVID
Database for annotation, visualization, and integrated discovery
FFPE
Formalin-fixed paraffin-embedded
FPKM
Fragments per kilobase per million mapped reads
FSGS
Focal segmental glomerulosclerosis
KEGG
Kyoto Encyclopedia of Genes and Genomes
NICD
Notch1 intracellular domain
PBS
Phosphate-buffered saline
PCV
Posterior crossvein
qRT-PCR
Quantitative reverse transcription polymerase chain reaction
SIOD
Schimke immuno-osseous dysplasia
SMARCAL1
SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily A-like 1

Background

Schimke immuno-osseous dysplasia (SIOD, OMIM 242900) is an autosomal recessive disease; its prominent features are facial dysmorphism, hyperpigmented macules, focal segmental glomerulosclerosis (FSGS), spondyloepiphyseal dysplasia, and T-cell immunodeficiency [13]. Additional features include hypothyroidism, abnormal dentition, bone marrow failure, thin hair, corneal opacities, arteriosclerosis, cerebral ischemia, and migraine-like headaches [25].
The renal disease begins as proteinuria, progresses to steroid-resistant nephropathy, and ultimately advances to end-stage renal disease [4, 6]. FSGS is the predominant renal pathology and is refractory to treatment with glucocorticoids, cyclosporine A, and cyclophosphamide [4, 6]. Suggesting a cell autonomous mechanism for the renal disease, renal transplantation is efficacious, and the disease does not recur in the graft [2, 4, 5].
Biallelic mutations of the SWI/SNF-related matrix-associated actin-dependent regulator of chromatin, subfamily A-like 1 (SMARCAL1) gene cause SIOD [7]. SMARCAL1 encodes a DNA annealing helicase that is a distant member of the SWI/SNF family of ATP-dependent chromatin remodeling proteins [8]. SMARCAL1 recognizes DNA structure, binds to open chromatin, is involved in the DNA damage response [9, 10] and DNA replication fork restart [11, 12], and, along with genetic and environmental factors, alters gene expression [13].
Gene expression changes appear critical to SIOD pathology. Full or partial explanations for the vascular disease and T-cell immunodeficiency of SIOD patients are respectively decreased expression of elastin (ELN) in the aorta [1416] and of interleukin 7 receptor alpha chain (IL7R) in the T cells [1719].
Based on these findings, we hypothesized that SMARCAL1 deficiency causes the renal disease of SIOD by altering gene expression. Studies of other glomerulopathies find increased Wnt [2023] and Notch signaling [2427] as causes of podocyte dysfunction. Canonical Wnt pathway activation proceeds via inhibition of β-catenin ubiquitination, saturation of the β-catenin destruction complex, cytoplasmic accumulation and nuclear translocation of newly synthesized unphosphorylated β-catenin, and subsequent activation of target gene transcription through interaction with transcription factors and transcriptional co-activators [28]. Notch pathway activation involves proteolytic cleavage of the Notch transmembrane receptor by an ADAM metalloproteinase and the γ-secretase complex, nuclear translocation of the released Notch1 intracellular domain (NICD), and subsequent activation of target gene transcription through interaction of the NICD with transcription factors and transcriptional co-activators [29]. Wnt and Notch signaling are critical for kidney development and become undetectable in the glomeruli of the postnatal kidney [26, 30].
Analyses presented herein showed upregulation of the Wnt and Notch signaling pathways in the SIOD kidney and genetic interaction between the Drosophila SMARCAL1 homologue and genes encoding components of the Wnt and Notch pathways. We suggest therefore that the upregulation of the Wnt and/or Notch pathways contributes to the renal disease in SIOD.

Methods

Patients and human tissues

The guardians of the patients referred to this study signed informed consent approved by the Research Ethics Board of the University of British Columbia (Vancouver, BC, Canada). Autopsy and biopsy tissues were obtained according to the protocol approved by the University of British Columbia (Vancouver, BC, Canada). The renal parameters and the SMARCAL1 mutations of the SIOD patients included in the study are listed in Table 1 and Additional file 1: Table S1, respectively.
Table 1
The renal parameters of the SIOD patients included in this study
Patient ID
Age at onset (years)
Age at death (years)
Nephrotic syndrome
Hypertension
Proteinuria
Hypercholesterolemia
Renal dialysis
Age at renal dialysis (years)
Renal transplant
Age at renal transplantation (years)
Renal pathology
SD4b
3
8
+
+
+
?
n/a
n/a
FSGS
SD26
<4
8
+
+
+
+
+
5
n/a
FSGS
SD60
7
13.7
+
+
+
?
+
12.5
+
13
FSGS
SD79
<4
10
+
+
n/a
n/a
FSGS
SD120
4.5
5.4
+
+
+
+
n/a
n/a
FSGS
SD121
2.5
4.8
+
+
+
n/a
n/a
Diffuse podocytopathy with early features of FSGS
SD131
3
4.6
+
+
+
+
+
3.8
n/a
Global glomerulosclerosis likely secondary to FSGS
SD146
2
4
+
+
n/a
n/a
FSGS
Abbreviations: + present, absent, ? unknown, FSGS focal segmental glomerulosclerosis, ID identification, n/a not applicable, SIOD Schimke immuno-osseous dysplasia
In accordance with institutional policies as approved by the Institutional Review Board (41557) at the University of Washington, human fetal kidney from second trimester elective terminations were provided as de-identified specimens by the Laboratory of Developmental Biology at the University of Washington (Seattle, WA), a National Institute of Child Health & Human Development supported program. De-identified control specimens provided according to the protocol H06-70283 approved by the Clinical Research Ethics Board at the University of British Columbia (Vancouver, BC, Canada) included renal biopsy sections from ten pediatric patients with isolated FSGS, postmortem kidney tissue from four pediatric patients, a skin biopsy from a 16-year-old female, and adenoma tissue from a 17-year-old female with familial adenomatous polyposis. Sample characteristics and use are summarized in Additional file 1: Table S2.

Drosophila melanogaster lines

The loss-of-function mutant Marcal1 del and the Marcal1 overexpression transgenic line pUAST-Marcal1/CyO; tubulin-GAL4/TM3, Sb 1 have been previously described [13] (Additional file 1: Figure S1). The C96-GAL4 UAS-Hrs/MKRS transgenic line, used to control for non-specific interactions with the GAL4-UAS system, was a gift from Dr. Hugo Bellen (Baylor College of Medicine, Houston, TX, USA). All other Drosophila stocks were obtained from the Bloomington Drosophila Stock Center (Bloomington, IN, USA).

RNA extraction

Total RNA was extracted from flash frozen kidney pulverized with a Bessman tissue pulverizer (Spectrum Laboratories, Rancho Dominguez, CA, USA) or from 8 Drosophila adult female flies of each genotype by using the RNeasy Mini Kit (Qiagen, Toronto, ON, Canada). Total RNA from formalin-fixed paraffin-embedded (FFPE) fetal kidney was isolated using the RNeasy FFPE Kit (Qiagen, Toronto, ON, Canada). Genomic DNA was removed by on-column DNase I digestion (Qiagen, Toronto, ON, Canada).

RNA-seq and KEGG pathway analysis

Strand-specific, paired-end RNA-seq on poly(A) RNA was performed by Macrogen (Seoul, Korea) using the TruSeq Stranded Total RNA Library Prep Kit (Illumina, San Diego, CA) and the HiSeq 2000 System (Illumina, San Diego, CA). This kit depleted the ribosomal RNA (rRNA) using Ribo-Zero rRNA reduction chemistry. Quantification was performed by calculating fragments per kilobase per million mapped reads (FPKM). Prior to fold change calculation and log2 transformation, a pseudocount of 1 was added to each FPKM value to reduce the inherent bias of finding gene expression changes in those genes where one sample has very little or no detectable gene expression [31]. The threshold for differential gene expression between the kidney from the SIOD patient and sex-matched unaffected control was set at log2 fold change (i.e., log2 (FPKMSIOD + 1/FPKMUNAFFECTED + 1)) > 1 or < −1. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis was performed with the online bioinformatic resource Database for Annotation, Visualization, and Integrated Discovery (DAVID) version 6.7 available at https://​david.​ncifcrf.​gov.

Reverse transcription

For total RNA extracted from flash frozen kidney, reverse transcription was performed with the RT2 First Strand Kit (Qiagen, Toronto, ON, Canada). For total RNA extracted from FFPE kidney or adult flies, reverse transcription was performed with the qScript cDNA SuperMix (Quanta Biosciences, Gaithersburg, MD, USA).

Gene expression arrays

The Wnt (PAHS-043Y) and Notch (PAHS-059Y) Signaling Pathway Plus PCR Arrays (Qiagen, Toronto, ON, Canada) and the RT2 Real-Time SYBR Green/Rox PCR Master Mix (Qiagen, Toronto, ON, Canada) were used to assess mRNA levels between the sex-matched unaffected control and the SIOD kidney according to the manufacturer’s specifications. The threshold for calling differential mRNA levels was a log2 fold change > 1 or < −1 and a p value of less than 0.05.

Quantitative PCR

SsoFast EvaGreen Supermix (Bio-Rad Laboratories, Mississauga, ON, Canada) was used with the StepOnePlus Real-Time PCR System (Applied Biosystems, Thermo Fisher Scientific, Waltham, MA, USA) for quantitative PCR. Human GAPDH and Drosophila Gapdh2 housekeeping genes were used as endogenous controls. The primer sequences used in this study are listed in Additional file 1: Table S3.

Indirect immunofluorescence

FFPE sections of tissue or cell pellets were cut at 5 microns. Following deparaffinization and rehydration, heat induced epitope retrieval was performed with sodium citrate buffer (10 mM sodium citrate, 0.05 % Tween 20, pH 6.0). Endogenous peroxidases were inactivated for 1 h at room temperature by incubating the sections with peroxidase quenching buffer (3 % hydrogen peroxide in 1× phosphate-buffered saline (PBS), 0.1 % Tween 20, pH 7.4 (PBSTw) for unphosphorylated β-catenin immunofluorescent staining or 1× PBS, 0.2 % Triton X-100, pH 7.4 (PBST) for the Notch1 intracellular domain (NICD) immunofluorescent staining). Non-specific protein binding was blocked by incubating the sections with blocking buffer (20 % normal goat serum, 10 % bovine serum albumin, 1× casein (Vector Laboratories, Burlington, ON, Canada) in PBSTw or PBST) overnight at 4 °C. Endogenous biotin, biotin receptors, and avidin binding sites were blocked with the Avidin/Biotin Blocking Kit (Vector Laboratories, Burlington, ON, Canada).
Rabbit anti-unphosphorylated β-catenin (clone D13A1, Cell Signaling Technology, Danvers, MA, USA) or rabbit anti-NICD (ab8925, Abcam, Toronto, ON, Canada) were used as primary antibodies. A biotinylated anti-rabbit IgG secondary antibody was used to detect the primary antibodies. Horseradish peroxidase-conjugated streptavidin was then used to detect the biotinylated anti-rabbit IgG secondary antibody. Subsequently, tyramide labeling was performed using Alexa Fluor 594 tyramide (Invitrogen, Thermo Fisher Scientific, Waltham, MA, USA). ProLong Gold Antifade Mountant with 4′, 6-diamidino-2-phenylindole (DAPI) (Invitrogen, Thermo Fisher Scientific, Waltham, MA, USA) was used to mount the sections and counterstain the DNA. Representative images were acquired using a 20×/0.75 Plan-APOCHROMAT, 40×/1.3 oil DIC Plan-NEOFLUAR, or 100×/1.30 oil Plan-NEOFLUAR objective lens on an Axiovert 200 inverted microscope, an AxioCam MR microscope camera, and the AxioVision software version 4.8 (Carl Zeiss, Toronto, ON, Canada). The glomerular β-catenin signal was quantified for each sample (see Additional file 1: Methods for further details).

Drosophila genetics studies

We performed an overexpression and loss-of-function genetic screen in Drosophila to determine whether the SMARCAL1 homologue Marcal1 genetically interacts with Wnt and Notch pathway genes (see Additional file 1: Methods for further details).

Statistics

For the KEGG pathway analysis, enrichment p values were corrected for multiple comparisons by the Bonferroni method. A p value of less than 0.05 was considered statistically significant. For the PCR expression arrays, data were analyzed by the 2-tailed Student’s t-test. A p value of less than 0.05 was considered statistically significant.

Results

Genome-wide gene expression analysis identifies increased mRNA levels of Wnt signaling pathway and target genes in an SIOD patient kidney

We hypothesized that SMARCAL1 deficiency leads to gene expression changes that contribute to the pathogenesis of the renal disease in SIOD. To test this, we used RNA-seq to compare the transcriptomes of kidney tissue from a 5.4-year-old male SIOD patient and a 3-year-old unaffected male. This comparison detected 2241 genes with increased mRNA levels (log2 fold change > 1) and 892 genes with decreased mRNA levels (log2 fold change < −1) in the SIOD kidney tissue. After Bonferroni correction, KEGG pathway analysis of the genes with decreased mRNA levels did not reveal any significantly enriched pathways. In contrast, KEGG pathway analysis of genes with increased mRNA levels revealed significantly enriched pathways of cellular adhesion (e.g., focal adhesion, cell adhesion molecules), immune function (e.g., leukocyte transendothelial migration, Fc gamma R-mediated phagocytosis), disease (e.g., systemic lupus erythematosus, pathways in cancer, colorectal cancer), and Wnt signaling (Fig. 1a and Additional file 1: Table S4).

Targeted gene expression analysis detects increased mRNA levels of Wnt and Notch signaling pathway and target genes in an SIOD patient kidney

Given that upregulation of the Wnt pathway [2023] or the Notch pathway [2427] is a cause of glomerulopathy, we measured mRNA levels of Wnt and Notch signaling pathway and target genes using the RT2 Profiler PCR Arrays. These analyses showed that of the 84 Wnt pathway-related genes tested, 30 were differentially expressed (Fig. 1b and Additional file 1: Table S5) and that of the 84 Notch pathway-related genes tested, 14 were differentially expressed (Fig. 1c and Additional file 1: Table S6). Wnt pathway-related genes with increased mRNA levels included ligands (e.g., WNT2B, WNT4, WNT6, WNT7A, WNT10A), components (e.g., AXIN2, FZD2, FZD7, SFRP1, SFRP4), and targets (e.g., AXIN2, CCND2, JUN, MMP7, MYC). Notch pathway-related genes with increased mRNA levels included components (e.g., DTX1) and targets (e.g., HEYL, IL2RA).

Markers of Wnt and Notch pathway activation are increased in the glomerular cells of postnatal SIOD patient kidneys comparable to isolated FSGS controls

Having established that several Wnt and Notch pathway-related genes and targets have altered expression in an SIOD kidney, we hypothesized that increased Wnt and Notch pathway signaling within the glomeruli contributes to the pathogenesis of FSGS in SIOD. To test this in additional SIOD patients, we used indirect immunofluorescence to profile the expression of unphosphorylated β-catenin and the nuclear localization of the Notch1 intracellular domain (NICD), which are respectively markers of canonical Wnt and Notch pathway activation [28, 29] (Additional file 1: Figure S2 and Fig. 3a). Compared to unaffected controls, most SIOD samples had increased glomerular staining for unphosphorylated β-catenin (6 of 7 patients) and nuclear NICD (6 of 8 patients) (Fig. 2, Fig. 3, Table 2, and Additional file 1: Figure S3). Similarly, most isolated FSGS samples had increased glomerular staining for unphosphorylated β-catenin (8 of 9 patients) and nuclear NICD (8 of 9 patients (Additional file 1: Figure S3, Figure S4, Figure S5, and Table 2).
Table 2
Summary of the β-catenin and NICD immunofluorescent analyses in SIOD and isolated FSGS patient kidney tissue
Patient ID
Unphosphorylated β-catenin expression
Nuclear NICD expression
SIOD patients
 SD4b
=
=
 SD26
 SD60
 SD60 Tx
=
=
 SD79
=
 SD120
 SD121
 SD131
n/aa
 SD146
Isolated FSGS patients
 FSGS-1
=
 FSGS-2
 FSGS-3
 FSGS-4
 FSGS-5
 FSGS-6
 FSGS-8
 FSGS-9
 FSGS-10
Abbreviation: = staining comparable to unaffected control kidney, increased staining compared to unaffected control kidney, FSGS focal segmental glomerulosclerosis, ID identification, n/a not available, NICD Notch1 intracellular domain, SIOD Schimke immuno-osseous dysplasia, Tx transplant
aNo more tissue sections were available for analysis

Markers of Wnt and Notch pathway activation are not increased a 15-week-gestation SMARCAL1-deficient kidney

To determine whether pathologically increased Wnt and Notch pathway signaling in SIOD begins prenatally, we performed indirect immunofluorescence for unphosphorylated β-catenin and NICD in a 15-week-gestation SMARCAL1-deficient kidney and age-matched unaffected kidneys. The SMARCAL1-deficient fetal kidney expressed comparable levels of unphosphorylated β-catenin and NICD to the age-matched controls in both S-shaped bodies and developing glomeruli (Additional file 1: Figure S6 and Figure S7). In agreement with these findings, expression analysis of several Wnt and Notch target genes in the SMARCAL1-deficient fetal kidney and age-matched controls demonstrated comparable expression levels (Additional file 1: Figure S8).

Markers of Wnt and Notch pathway activation are not increased in the transplanted kidney of an SIOD patient

Our previous studies have shown that the renal disease of SIOD is cell autonomous [5, 32]; therefore, we hypothesized that if the increased glomerular levels of unphosphorylated β-catenin and NICD are potentially causative of the renal disease in SIOD, then the levels of unphosphorylated β-catenin and NICD are not increased in renal grafts of SIOD patients. To test this hypothesis, we performed indirect immunofluorescence for unphosphorylated β-catenin and NICD in the transplanted kidney of an SIOD patient and observed a staining pattern and intensity similar to that of unaffected controls for unphosphorylated β-catenin and NICD (Additional file 1: Figure S3, Figure S4, and Figure S5).

Drosophila Marcal1 genetically interacts with the Wnt and Notch signaling pathways

To assess whether the upregulation of the Wnt and Notch signaling pathways is a genetic consequence of SMARCAL1 deficiency and not simply an end product of the tissue pathology, we performed overexpression and loss-of-function genetic screens in Drosophila. By assessing the suppression or enhancement of ectopic wing veins induced by Marcal1 overexpression [13], we found that both Wnt and Notch pathway genes genetically interacted with Marcal1 (Additional file 1: Table S7, Table S8, Figure S9, Figure S10, and Figure S11).
To confirm these interactions, we performed the reciprocal analysis, i.e., analysis of the suppression or enhancement of phenotypes associated with Wnt and Notch pathway mutants. For the well-characterized wing, eye, and bristle phenotypes of Notch pathway mutants, Marcal1 loss and gain suppressed or enhanced phenotypes for Notch (N) mutants, Delta (Dl) mutants, Hairless (H) mutants, and a fringe (fng) mutant (Fig. 4a and b, Additional file 1: Table S9 and Figure S12). No genetic interaction was observed between Marcal1 loss or gain and a Serrate (Ser) mutant (Fig. 4a and Additional file 1: Table S9).

Discussion

Herein we identify increased signaling of the Wnt and Notch pathways as potential causes for the renal disease in SIOD. Most SIOD kidneys exhibited increased levels of unphosphorylated β-catenin and NICD respectively indicating increased Wnt and Notch pathway activity. Similarly, most isolated FSGS kidneys had upregulated unphosphorylated β-catenin and NICD. The failure to observe increased unphosphorylated β-catenin and NICD in the renal graft of an SIOD patient suggests that these molecular findings are inherent to the diseased kidney and not induced from outside of the kidney. The genetic interaction between Marcal1 and the Wnt and Notch pathway genes in Drosophila suggests that that the altered signaling of these pathways is a direct or indirect consequence of SMARCAL1 deficiency.
The consistency of increased markers for both activation of the Wnt and Notch pathways in both SIOD and isolated FSGS control kidneys suggests that activation of both pathways underlie the renal disease of SIOD and isolated FSGS (Fig. 4c). Activation of both pathways is not essential for induction of SIOD renal disease or isolated FSGS, however, because a few samples showed activation of only one or neither of these pathways (Fig. 4c).
Based on our observations in the 15-week-gestation fetal kidney, the potentially pathological activation of Wnt and Notch signaling in the SIOD kidneys appears to arise after this stage of renal development. Further studies are required to define precisely the timing of the pathological activation of these pathways.
Although the Notch pathway gene expression changes were not identified in the KEGG pathway analysis of the transcriptome, the high level of crosstalk between the Wnt and Notch signaling pathways [33], and their role in kidney development and disease prompted us to also investigate the upregulation of the Notch pathway as a potential cause for the FSGS in SIOD. Possible reasons for the transcriptome analysis not detecting the upregulation of the Notch pathway include pathway size bias inherent to KEGG pathway analysis (the Wnt signaling pathway includes 141 genes, whereas the Notch signaling pathway includes 48 genes) and tissue heterogeneity.
The mechanism by which SMARCAL1 deficiency gives rise to tissue-specific changes in gene expression is incompletely understood. It could arise from a direct consequence of SMARCAL1 deficiency on the DNA structure of a gene or of the genes encoding the transcriptional regulators of that gene. Consistent with this, we previously observed that SMARCAL1 homologues bind transcriptionally active chromatin and modulate gene expression [13]. Sharma et al. (2015) recently showed that the bovine orthologue of SMARCAL1 negatively and directly regulates the transcription of MYC by altering the conformation of its promoter [34]. Alternatively, because stalled replication forks induce epigenetic changes that alter gene expression [35, 36], impedance of DNA replication fork restart by SMARCAL1 deficiency might contribute to the changes in gene expression. Consistent with the latter possibility, we recently observed hypermethylation of the IL7R promoter in the T cells of SIOD patients [19]; reduced IL7R expression in human CD8+ T cells is associated with hypermethylation of the IL7R promoter [37].
A limitation of the study was the use of whole kidney to profile differential gene expression in an SIOD kidney. Given that the primary lesion is limited to the glomeruli, the affected tissue represents a small fraction of the total tissue. Although several human gene expression studies on FSGS have used isolated glomeruli [38, 39], others have successfully used renal biopsies [40]. Similar to other human gene expression studies of FSGS [3840], the expression of podocyte-specific genes including NPHS1, NPHS2, and WT1 were downregulated in the SIOD kidney, and most of the KEGG pathways that were enriched in our list of upregulated genes were also enriched in the prior studies, including the Wnt signaling pathway [38].
A second limitation of the study was that only unphosphorylated β-catenin and nuclear NICD were examined by immunofluorescence as measures of pathway activation. This constraint arose secondary to limited tissue. We selected these proteins because they are the primary effectors of and activation markers for the canonical Wnt and Notch signaling pathways. However, Wnt signaling has canonical and non-canonical pathways, and there is also Wnt-independent β-catenin activation [41]. Notch signaling also has canonical and non-canonical pathways as well as three Notch receptors in addition to Notch1 [42]. Our findings nonetheless set a precedent for future studies examining the pathogenesis of renal disease in SIOD.

Conclusions

In summary, our findings show that the Wnt and Notch pathways are upregulated in the SIOD patient kidney and that Marcal1, the Drosophila SMARCAL1 homologue, genetically interacts with Wnt and Notch pathway genes. Based on these findings, the renal disease of SIOD is yet another clinically distinctive feature of SIOD likely arising through alterations of gene expression.

Acknowledgements

We are grateful to all of the patients and family members who have contributed to this study. The authors thank Theresa Sturby (Children’s and Women’s Health Centre of British Columbia) for her technical expertise, and Drs. Darren Bridgewater and Alireza Barandaran-Heravi for critical review of this manuscript.

Funding

This work was supported in part by the New Investigator Award jointly sponsored by the SickKids Foundation and the Canadian Institutes of Health Research Institute of Human Development, Child and Youth Health (XG09-025 to CFB); the Michael Smith Foundation for Health Research (CI-SCH-O1899(07–1) to CFB); The Little Giants Foundation (CFB); and the Asociacion Española de Displasias Oseas Minoritarias (CFB). Human fetal tissue was obtained through the Laboratory of Developmental Biology project supported by the National Institutes of Health Award Number 5R24HD000836 from the Eunice Kennedy Shriver National Institute of Child Health & Human Development. MM was supported by a Four Year Doctoral Fellowship from the University of British Columbia. CFB is a scholar of the Michael Smith Foundation for Health Research and a Clinical Investigator of the Child & Family Research Institute.

Availability of data and materials

The dataset supporting the conclusions of this article is available in the Gene Expression Omnibus (GEO) repository, Series record GSE75061.

Authors’ contributions

All authors have made substantial contributions to the article by participating in the conception and design (MM, CFB), acquisition of data (MM, CM, KB, KC, YA, AB, DB, MB, JC, EC, AD, GD, M Gentile, M Giordano, AKG, RG, MJ, KK, E Lerut, E Levtchenko, LM, CM, BN, DP, JS, PS, UY, ZY, JZ, GH, CFB), analysis and interpretation of data (MM, CM, KB, KC, BN, CFB), drafting the manuscript (MM, CFB), or revising it critically for important intellectual content (MM, CM, KB, KC, YA, AB, DB, MB, JC, EC, AD, GD, MGentile, MGiordano, AKG, RG, MJ, KK, ELerut, ELevtchenko, LM, CM, BN, DP, JS, PS, UY, ZY, JZ, GH, CFB). All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
Not applicable.
The guardians of the patients referred to this study signed informed consent approved by the Institutional Review Board of the University of British Columbia (Vancouver, BC, Canada). Autopsy and biopsy tissues were obtained according to the protocol approved by the University of British Columbia (Vancouver, BC, Canada).
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.
Literatur
1.
Zurück zum Zitat Schimke RN, Horton WA, King CR. Chondroitin-6-sulphaturia, defective cellular immunity, and nephrotic syndrome. Lancet. 1971;2:1088–9.CrossRefPubMed Schimke RN, Horton WA, King CR. Chondroitin-6-sulphaturia, defective cellular immunity, and nephrotic syndrome. Lancet. 1971;2:1088–9.CrossRefPubMed
2.
Zurück zum Zitat Ehrich JH, Burchert W, Schirg E, Krull F, Offner G, Hoyer PF, et al. Steroid resistant nephrotic syndrome associated with spondyloepiphyseal dysplasia, transient ischemic attacks and lymphopenia. Clin Nephrol. 1995;43:89–95.PubMed Ehrich JH, Burchert W, Schirg E, Krull F, Offner G, Hoyer PF, et al. Steroid resistant nephrotic syndrome associated with spondyloepiphyseal dysplasia, transient ischemic attacks and lymphopenia. Clin Nephrol. 1995;43:89–95.PubMed
3.
Zurück zum Zitat Spranger J, Hinkel GK, Stoss H, Thoenes W, Wargowski D, Zepp F. Schimke immuno-osseous dysplasia: a newly recognized multisystem disease. J Pediatr. 1991;119:64–72.CrossRefPubMed Spranger J, Hinkel GK, Stoss H, Thoenes W, Wargowski D, Zepp F. Schimke immuno-osseous dysplasia: a newly recognized multisystem disease. J Pediatr. 1991;119:64–72.CrossRefPubMed
4.
Zurück zum Zitat Boerkoel CF, O’Neill S, Andre JL, Benke PJ, Bogdanovic R, Bulla M, et al. Manifestations and treatment of Schimke immuno-osseous dysplasia: 14 new cases and a review of the literature. Eur J Pediatr. 2000;159:1–7.CrossRefPubMed Boerkoel CF, O’Neill S, Andre JL, Benke PJ, Bogdanovic R, Bulla M, et al. Manifestations and treatment of Schimke immuno-osseous dysplasia: 14 new cases and a review of the literature. Eur J Pediatr. 2000;159:1–7.CrossRefPubMed
5.
Zurück zum Zitat Clewing JM, Antalfy BC, Lücke T, Najafian B, Marwedel KM, Hori A, et al. Schimke immuno-osseous dysplasia: a clinicopathological correlation. J Med Genet. 2007;44:122–30.CrossRefPubMedPubMedCentral Clewing JM, Antalfy BC, Lücke T, Najafian B, Marwedel KM, Hori A, et al. Schimke immuno-osseous dysplasia: a clinicopathological correlation. J Med Genet. 2007;44:122–30.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Sarin S, Javidan A, Boivin F, Alexopoulou I, Lukic D, Svajger B, et al. Insights into the renal pathogenesis in schimke immuno-osseous dysplasia: a renal histological characterization and expression analysis. J Histochem Cytochem. 2015;63:32–44.CrossRefPubMedPubMedCentral Sarin S, Javidan A, Boivin F, Alexopoulou I, Lukic D, Svajger B, et al. Insights into the renal pathogenesis in schimke immuno-osseous dysplasia: a renal histological characterization and expression analysis. J Histochem Cytochem. 2015;63:32–44.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Boerkoel CF, Takashima H, John J, Yan J, Stankiewicz P, Rosenbarker L, et al. Mutant chromatin remodeling protein SMARCAL1 causes Schimke immuno-osseous dysplasia. Nat Genet. 2002;30:215–20.CrossRefPubMed Boerkoel CF, Takashima H, John J, Yan J, Stankiewicz P, Rosenbarker L, et al. Mutant chromatin remodeling protein SMARCAL1 causes Schimke immuno-osseous dysplasia. Nat Genet. 2002;30:215–20.CrossRefPubMed
9.
10.
Zurück zum Zitat Yusufzai T, Kong X, Yokomori K, Kadonaga JT. The annealing helicase HARP is recruited to DNA repair sites via an interaction with RPA. Genes Dev. 2009;23:2400–4.CrossRefPubMedPubMedCentral Yusufzai T, Kong X, Yokomori K, Kadonaga JT. The annealing helicase HARP is recruited to DNA repair sites via an interaction with RPA. Genes Dev. 2009;23:2400–4.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Bansbach CE, Betous R, Lovejoy CA, Glick GG, Cortez D. The annealing helicase SMARCAL1 maintains genome integrity at stalled replication forks. Genes Dev. 2009;23:2405–14.CrossRefPubMedPubMedCentral Bansbach CE, Betous R, Lovejoy CA, Glick GG, Cortez D. The annealing helicase SMARCAL1 maintains genome integrity at stalled replication forks. Genes Dev. 2009;23:2405–14.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Baradaran-Heravi A, Cho KS, Tolhuis B, Sanyal M, Morozova O, Morimoto M, et al. Penetrance of biallelic SMARCAL1 mutations is associated with environmental and genetic disturbances of gene expression. Hum Mol Genet. 2012;21:2572–87.CrossRefPubMedPubMedCentral Baradaran-Heravi A, Cho KS, Tolhuis B, Sanyal M, Morozova O, Morimoto M, et al. Penetrance of biallelic SMARCAL1 mutations is associated with environmental and genetic disturbances of gene expression. Hum Mol Genet. 2012;21:2572–87.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Curran ME, Atkinson DL, Ewart AK, Morris CA, Leppert MF, Keating MT. The elastin gene is disrupted by a translocation associated with supravalvular aortic stenosis. Cell. 1993;73:159–68.CrossRefPubMed Curran ME, Atkinson DL, Ewart AK, Morris CA, Leppert MF, Keating MT. The elastin gene is disrupted by a translocation associated with supravalvular aortic stenosis. Cell. 1993;73:159–68.CrossRefPubMed
15.
Zurück zum Zitat Li DY, Brooke B, Davis EC, Mecham RP, Sorensen LK, Boak BB, et al. Elastin is an essential determinant of arterial morphogenesis. Nature. 1998;393:276–80.CrossRefPubMed Li DY, Brooke B, Davis EC, Mecham RP, Sorensen LK, Boak BB, et al. Elastin is an essential determinant of arterial morphogenesis. Nature. 1998;393:276–80.CrossRefPubMed
16.
Zurück zum Zitat Morimoto M, Yu Z, Stenzel P, Clewing JM, Najafian B, Mayfield C, et al. Reduced elastogenesis: a clue to the arteriosclerosis and emphysematous changes in schimke immuno-osseous dysplasia? Orphanet J Rare Dis. 2012;7:70.CrossRefPubMedPubMedCentral Morimoto M, Yu Z, Stenzel P, Clewing JM, Najafian B, Mayfield C, et al. Reduced elastogenesis: a clue to the arteriosclerosis and emphysematous changes in schimke immuno-osseous dysplasia? Orphanet J Rare Dis. 2012;7:70.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Puel A, Ziegler SF, Buckley RH, Leonard WJ. Defective IL7R expression in T(−)B(+)NK(+) severe combined immunodeficiency. Nat Genet. 1998;20:394–7.CrossRefPubMed Puel A, Ziegler SF, Buckley RH, Leonard WJ. Defective IL7R expression in T(−)B(+)NK(+) severe combined immunodeficiency. Nat Genet. 1998;20:394–7.CrossRefPubMed
18.
Zurück zum Zitat Roifman CM, Zhang J, Chitayat D, Sharfe N. A partial deficiency of interleukin-7R alpha is sufficient to abrogate T-cell development and cause severe combined immunodeficiency. Blood. 2000;96:2803–7.PubMed Roifman CM, Zhang J, Chitayat D, Sharfe N. A partial deficiency of interleukin-7R alpha is sufficient to abrogate T-cell development and cause severe combined immunodeficiency. Blood. 2000;96:2803–7.PubMed
19.
Zurück zum Zitat Sanyal M, Morimoto M, Baradaran-Heravi A, Choi K, Kambham N, Jensen K, et al. Lack of IL7Ralpha expression in T cells is a hallmark of T-cell immunodeficiency in Schimke immuno-osseous dysplasia (SIOD). Clin Immunol. 2015;161:355–65.CrossRefPubMed Sanyal M, Morimoto M, Baradaran-Heravi A, Choi K, Kambham N, Jensen K, et al. Lack of IL7Ralpha expression in T cells is a hallmark of T-cell immunodeficiency in Schimke immuno-osseous dysplasia (SIOD). Clin Immunol. 2015;161:355–65.CrossRefPubMed
20.
Zurück zum Zitat Dai C, Stolz DB, Kiss LP, Monga SP, Holzman LB, Liu Y. Wnt/beta-catenin signaling promotes podocyte dysfunction and albuminuria. J Am Soc Nephrol. 2009;20:1997–2008.CrossRefPubMedPubMedCentral Dai C, Stolz DB, Kiss LP, Monga SP, Holzman LB, Liu Y. Wnt/beta-catenin signaling promotes podocyte dysfunction and albuminuria. J Am Soc Nephrol. 2009;20:1997–2008.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Kato H, Gruenwald A, Suh JH, Miner JH, Barisoni-Thomas L, Taketo MM, et al. Wnt/beta-catenin pathway in podocytes integrates cell adhesion, differentiation, and survival. J Biol Chem. 2011;286:26003–15.CrossRefPubMedPubMedCentral Kato H, Gruenwald A, Suh JH, Miner JH, Barisoni-Thomas L, Taketo MM, et al. Wnt/beta-catenin pathway in podocytes integrates cell adhesion, differentiation, and survival. J Biol Chem. 2011;286:26003–15.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Shkreli M, Sarin KY, Pech MF, Papeta N, Chang W, Brockman SA, et al. Reversible cell-cycle entry in adult kidney podocytes through regulated control of telomerase and Wnt signaling. Nat Med. 2012;18:111–9.CrossRef Shkreli M, Sarin KY, Pech MF, Papeta N, Chang W, Brockman SA, et al. Reversible cell-cycle entry in adult kidney podocytes through regulated control of telomerase and Wnt signaling. Nat Med. 2012;18:111–9.CrossRef
23.
Zurück zum Zitat He W, Tan RJ, Li Y, Wang D, Nie J, Hou FF, et al. Matrix metalloproteinase-7 as a surrogate marker predicts renal Wnt/beta-catenin activity in CKD. J Am Soc Nephrol. 2012;23:294–304.CrossRefPubMedPubMedCentral He W, Tan RJ, Li Y, Wang D, Nie J, Hou FF, et al. Matrix metalloproteinase-7 as a surrogate marker predicts renal Wnt/beta-catenin activity in CKD. J Am Soc Nephrol. 2012;23:294–304.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Niranjan T, Bielesz B, Gruenwald A, Ponda MP, Kopp JB, Thomas DB, et al. The Notch pathway in podocytes plays a role in the development of glomerular disease. Nat Med. 2008;14:290–8.CrossRefPubMed Niranjan T, Bielesz B, Gruenwald A, Ponda MP, Kopp JB, Thomas DB, et al. The Notch pathway in podocytes plays a role in the development of glomerular disease. Nat Med. 2008;14:290–8.CrossRefPubMed
25.
Zurück zum Zitat Waters AM, Wu MY, Onay T, Scutaru J, Liu J, Lobe CG, et al. Ectopic notch activation in developing podocytes causes glomerulosclerosis. J Am Soc Nephrol. 2008;19:1139–57.CrossRefPubMedPubMedCentral Waters AM, Wu MY, Onay T, Scutaru J, Liu J, Lobe CG, et al. Ectopic notch activation in developing podocytes causes glomerulosclerosis. J Am Soc Nephrol. 2008;19:1139–57.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Lasagni L, Ballerini L, Angelotti ML, Parente E, Sagrinati C, Mazzinghi B, et al. Notch activation differentially regulates renal progenitors proliferation and differentiation toward the podocyte lineage in glomerular disorders. Stem Cells. 2010;28:1674–85.CrossRefPubMedPubMedCentral Lasagni L, Ballerini L, Angelotti ML, Parente E, Sagrinati C, Mazzinghi B, et al. Notch activation differentially regulates renal progenitors proliferation and differentiation toward the podocyte lineage in glomerular disorders. Stem Cells. 2010;28:1674–85.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Murea M, Park JK, Sharma S, Kato H, Gruenwald A, Niranjan T, et al. Expression of Notch pathway proteins correlates with albuminuria, glomerulosclerosis, and renal function. Kidney Int. 2010;78:514–22.CrossRefPubMed Murea M, Park JK, Sharma S, Kato H, Gruenwald A, Niranjan T, et al. Expression of Notch pathway proteins correlates with albuminuria, glomerulosclerosis, and renal function. Kidney Int. 2010;78:514–22.CrossRefPubMed
28.
29.
Zurück zum Zitat Bray SJ. Notch signalling: a simple pathway becomes complex. Nat Rev Mol Cell Biol. 2006;7:678–89.CrossRefPubMed Bray SJ. Notch signalling: a simple pathway becomes complex. Nat Rev Mol Cell Biol. 2006;7:678–89.CrossRefPubMed
31.
Zurück zum Zitat Warden CD, Yuan Y-C, Wu X. Optimal calculation of RNA-seq fold-change values. Int J Comput Bioinfo In Silico Model. 2013;2:285–92. Warden CD, Yuan Y-C, Wu X. Optimal calculation of RNA-seq fold-change values. Int J Comput Bioinfo In Silico Model. 2013;2:285–92.
32.
Zurück zum Zitat Lücke T, Marwedel KM, Kanzelmeyer NK, Hori A, Offner G, Kreipe HH, et al. Generalized atherosclerosis sparing the transplanted kidney in Schimke disease. Pediatr Nephrol. 2004;19:672–5.CrossRefPubMed Lücke T, Marwedel KM, Kanzelmeyer NK, Hori A, Offner G, Kreipe HH, et al. Generalized atherosclerosis sparing the transplanted kidney in Schimke disease. Pediatr Nephrol. 2004;19:672–5.CrossRefPubMed
33.
Zurück zum Zitat Collu GM, Hidalgo-Sastre A, Brennan K. Wnt-Notch signalling crosstalk in development and disease. Cell Mol Life Sci. 2014;71:3553–67.CrossRefPubMed Collu GM, Hidalgo-Sastre A, Brennan K. Wnt-Notch signalling crosstalk in development and disease. Cell Mol Life Sci. 2014;71:3553–67.CrossRefPubMed
34.
Zurück zum Zitat Sharma T, Bansal R, Haokip DT, Goel I, Muthuswami R. SMARCAL1 negatively regulates c-Myc transcription by altering the conformation of the promoter region. Sci Rep. 2015;5:17910.CrossRefPubMedPubMedCentral Sharma T, Bansal R, Haokip DT, Goel I, Muthuswami R. SMARCAL1 negatively regulates c-Myc transcription by altering the conformation of the promoter region. Sci Rep. 2015;5:17910.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Schiavone D, Guilbaud G, Murat P, Papadopoulou C, Sarkies P, Prioleau MN, et al. Determinants of G quadruplex-induced epigenetic instability in REV1-deficient cells. EMBO J. 2014;33:2507–20.CrossRefPubMedPubMedCentral Schiavone D, Guilbaud G, Murat P, Papadopoulou C, Sarkies P, Prioleau MN, et al. Determinants of G quadruplex-induced epigenetic instability in REV1-deficient cells. EMBO J. 2014;33:2507–20.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Khurana S, Oberdoerffer P. Replication stress: a lifetime of epigenetic change. Genes (Basel). 2015;6:858–77. Khurana S, Oberdoerffer P. Replication stress: a lifetime of epigenetic change. Genes (Basel). 2015;6:858–77.
37.
Zurück zum Zitat Kim HR, Hwang KA, Kim KC, Kang I. Down-regulation of IL-7Ralpha expression in human T cells via DNA methylation. J Immunol. 2007;178:5473–9.CrossRefPubMed Kim HR, Hwang KA, Kim KC, Kang I. Down-regulation of IL-7Ralpha expression in human T cells via DNA methylation. J Immunol. 2007;178:5473–9.CrossRefPubMed
38.
Zurück zum Zitat Bennett MR, Czech KA, Arend LJ, Witte DP, Devarajan P, Potter SS. Laser capture microdissection-microarray analysis of focal segmental glomerulosclerosis glomeruli. Nephron Exp Nephrol. 2007;107:e30–40.CrossRefPubMed Bennett MR, Czech KA, Arend LJ, Witte DP, Devarajan P, Potter SS. Laser capture microdissection-microarray analysis of focal segmental glomerulosclerosis glomeruli. Nephron Exp Nephrol. 2007;107:e30–40.CrossRefPubMed
39.
Zurück zum Zitat Hodgin JB, Borczuk AC, Nasr SH, Markowitz GS, Nair V, Martini S, et al. A molecular profile of focal segmental glomerulosclerosis from formalin-fixed, paraffin-embedded tissue. Am J Pathol. 2010;177:1674–86.CrossRefPubMedPubMedCentral Hodgin JB, Borczuk AC, Nasr SH, Markowitz GS, Nair V, Martini S, et al. A molecular profile of focal segmental glomerulosclerosis from formalin-fixed, paraffin-embedded tissue. Am J Pathol. 2010;177:1674–86.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Schwab K, Witte DP, Aronow BJ, Devarajan P, Potter SS, Patterson LT. Microarray analysis of focal segmental glomerulosclerosis. Am J Nephrol. 2004;24:438–47.CrossRefPubMed Schwab K, Witte DP, Aronow BJ, Devarajan P, Potter SS, Patterson LT. Microarray analysis of focal segmental glomerulosclerosis. Am J Nephrol. 2004;24:438–47.CrossRefPubMed
41.
Zurück zum Zitat Haq S, Michael A, Andreucci M, Bhattacharya K, Dotto P, Walters B, et al. Stabilization of beta-catenin by a Wnt-independent mechanism regulates cardiomyocyte growth. Proc Natl Acad Sci U S A. 2003;100:4610–5.CrossRefPubMedPubMedCentral Haq S, Michael A, Andreucci M, Bhattacharya K, Dotto P, Walters B, et al. Stabilization of beta-catenin by a Wnt-independent mechanism regulates cardiomyocyte growth. Proc Natl Acad Sci U S A. 2003;100:4610–5.CrossRefPubMedPubMedCentral
Metadaten
Titel
Increased Wnt and Notch signaling: a clue to the renal disease in Schimke immuno-osseous dysplasia?
verfasst von
Marie Morimoto
Clara Myung
Kimberly Beirnes
Kunho Choi
Yumi Asakura
Arend Bokenkamp
Dominique Bonneau
Milena Brugnara
Joel Charrow
Estelle Colin
Amira Davis
Georges Deschenes
Mattia Gentile
Mario Giordano
Andrew K. Gormley
Rajeshree Govender
Mark Joseph
Kory Keller
Evelyne Lerut
Elena Levtchenko
Laura Massella
Christy Mayfield
Behzad Najafian
David Parham
Jurgen Spranger
Peter Stenzel
Uluc Yis
Zhongxin Yu
Jonathan Zonana
Glenda Hendson
Cornelius F. Boerkoel
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Orphanet Journal of Rare Diseases / Ausgabe 1/2016
Elektronische ISSN: 1750-1172
DOI
https://doi.org/10.1186/s13023-016-0519-7

Weitere Artikel der Ausgabe 1/2016

Orphanet Journal of Rare Diseases 1/2016 Zur Ausgabe