Skip to main content
Erschienen in: Pediatric Nephrology 5/2007

Open Access 01.05.2007 | Educational Feature

Genetics of focal segmental glomerulosclerosis

verfasst von: Robert P. Woroniecki, Jeffrey B. Kopp

Erschienen in: Pediatric Nephrology | Ausgabe 5/2007

Abstract

The recent advances in understanding the pathophysiology of focal segmental glomerulosclerosis (FSGS) and molecular function of glomerular filtration barrier come directly from genetic linkage and positional cloning studies. The exact role and function of the newly discovered genes and proteins are being investigated by in vitro and in vivo mechanistic studies. Those genes and proteins interactions seem to change susceptibility to kidney disease progression. Better understanding of their exact role in the development of FSGS may influence future therapies and outcomes in this complex disease.

Introduction

Focal segmental glomerulosclerosis (FSGS) is not a single disease, as initially thought [1], but the histological expression of a variety of distinct conditions [2]. It is a clinically and genetically heterogeneous entity characterized by a common renal biopsy picture of segmental scarring of the glomerular capillary tuft affecting one or more glomeruli. Immunoglobulin (Ig)M and complement C3 may be present in mesangium, attributed to macromolecular trapping, but other immune deposits are absent. Hyaline deposits may occupy capillary loops, initially in a subendothelial location. In general, FSGS can be divided into three etiologic categories: idiopathic, genetic, and reactive (the latter including postadaptive and medication-associated forms). Although the abnormalities of glomerular filtration barrier have been implicated in the pathophysiology of nephrotic syndrome for the last 30–40 years, it was only during the last decade when genetic linkage studies coupled with positional cloning uncovered new genes and their products that were causally linked to human nephrotic syndrome. Identifying expression abnormalities of those genes contributed to the understanding of the pathophysiology of FSGS in recent years.
Understanding the genetics of FSGS begins with appreciating the molecular composition of glomerular filtration barrier comprised of podocytes, basement membrane (GBM), and fenestrated endothelium. This barrier separates blood from the urinary space and under physiologic conditions selectively permits the ultrafiltration of solutes and preventing the excessive leakage of large molecules, such as albumin and clotting factors, with a molecular weight greater than 40 kDa [3]. The glomerular filtration barrier becomes incompetent and “leaky” in the case of nephrotic syndrome, including FSGS. Under physiologic conditions, podocytes are terminally differentiated epithelial cells with a cell body and several foot processes that are in contact with each other through the interpodocyte connection, called slit diaphragm (SD). All of the genetic defects identified to date affect gene transcription or assembly of critical podocyte functional structures, including SD, actin-based cytoskeleton, and adhesion complexes. These genes include nephrin (NPHS1), podocin (NPHS2), alpha-actinin-4 (ACTN-4), CD2-associated protein (CD2AP), Wilm’s tumor gene (WT1), and transient receptor potential cation 6 (TRPC6) (Table 1). Recently, mutations in phospholipase epsilon C have been identified as a cause of steroid-sensitive nephrotic syndrome [4]. It is still not understood why the FSGS lesion is focal (only some glomeruli are affected) and segmental (only part of the glomerulus has the anatomic lesion).
Table 1
Gene mutations that are causally linked to focal segmental glomerulosclerosis (FSGS)
Gene symbol
Gene locus
Protein
Mode of inheritance
Renal manifestations
Extrarenal manifestations
NPHS2
1q25.31
Podocin
Autosomal recessive
Minimal change nephropathy, FSGS
None
ACTN4
19q13
Alpha-actinin-4
Autosomal dominant
FSGS
None
TRPC6
11q21.22
Transient receptor potential cation channel 6
Autosomal dominant
FSGS
None
PLCE1
10q23.24
Phospholipase C epsilon
Autosomal recessive
Diffuse mesangial sclerosis and FSGS
None
WT1
11p13
Wilm’s tumor suppressor protein
Autosomal dominant, de novo mutation
Diffuse mesangial sclerosis and FSGS
Genitourinary abnormalities
LMXB1
9q34.1
Lim homeobox transcription factor 1β
Autosomal dominant
FSGS
Dystrophic nails, absent or malformed patella
tRNALeu
Mitochondrial genome
NA
Maternal
FSGS, tubulointerstitial nephritis
Muscle and brain disease, lactic acidosis, deafness, diabetes mellitus
COQ2
4q21.22
Coenzyme Q2 homolog, prenyltransferase
Autosomal recessive
FSGS
Neurologic and muscle abnormalities
ITGB4
17q25.1
Integrin β4
Autosomal recessive
FSGS
Epidermolysis bullosa

Nephrin (NPHS1)

The NPHS1 gene mutations cause congenital nephrotic syndrome of the Finnish type, an autosomal recessive disorder characterized by massive proteinuria in utero and nephrosis at birth. Although this disease is not pathologically characterized by FSGS, it was the first mutation of structural protein of podocyte causally linked to nephrotic syndrome [5]. Moreover, nephrin deficiency was detected in other forms of nephrotic syndrome, and an overlap in the genes encoding nephrin/podocin (NPHS1/NPHS2) mutation spectrum (a triallelic hit) has recently been documented in patients with congenital FSGS [6]. NPHS1 product nephrin has 1,241 amino acid residues and belongs to the immunoglobulin (Ig) family of cell adhesion molecules. It contains a transmembrane domain, eight Ig-like repeats, and one fibronectin III-like module. Nephrin was localized by immunogold labeling and electron tomography at the slit between podocyte foot processes in a network of 35-nm long, globular cross-strands lining lateral, elongated pores in a zipper-like model [7]. Those pores are the same as or smaller than albumin molecules. Patients with NPHS1 mutations and nephrin-knockout mice had only narrow filtration slits that lacked the slit diaphragm network and the 35-nm-long strands; these findings suggest that nephrin molecules are directly involved in constituting the macromolecule-retaining slit diaphragm and its pores [8]. Functionally, nephrin controls the podocyte cytoskeleton in vivo by interacting with other proteins, such as Src homology-2 (SH2)/SH3 domain-containing Nck adaptor proteins [7]. Originally described mutation in Finnish patients was a two-nucleotide deletion in exon 2 [5], but other mutations have been reported since. Those include deletions, insertions, nonsense, missense, splicing mutations, and common polymorphisms [911].

Podocin (NPHS2)

Positional cloning identified NPHS2, encoding podocin, as a causative gene in autosomal recessive steroid-resistant nephrotic syndrome, including FSGS [12]. NPHS2 encodes a putative 383 amino acid protein of approximately 42 kDa. It belongs to the stomatin family proteins (band-7 proteins). NPHS2 expression is restricted to the podocytes, as shown by in situ hybridization studies, and the protein is an integral plasma membrane protein associated with specialized lipid raft microdomains [12]. Podocin is required for the recruitment of nephrin into lipid rafts; it interacts with nephrin and CD2AP [13] and facilitates nephrin signaling [14]. In podocin-null mice, nephrin expression is increased, whereas ZO1 (tight junction protein) and CD2AP expression is decreased. The mice die a few days after birth from renal failure caused by diffuse mesangial sclerosis, and they develop proteinuria during the antenatal period [15].
At present, at least 26 different NPHS2 mutations associated with FSGS have been described, with a mutation defined as a polymorphism that is present in homozygosity in at least one patient with FSGS [12, 1620]. Many patients are compound heterozygotes with two distinct mutations. The age of onset is generally in infancy or childhood, but a few cases of adult onset kidney disease have been described, with the oldest patient presenting at age 36. In populations of European ancestry, NPHS2 mutations are present in 26% of families with familial FSGS and 12–19% of sporadic pediatric FSGS [16, 18]. On the other hand, NPHS2 mutations associated with pediatric FSGS appear to be uncommon in children of Asian ancestry [21]; little is known about children of African ancestry. FSGS associated with NPHS2 mutations is uniformly steroid resistant and generally shows poor response to cyclosporine as well [18]. Although early reports suggested otherwise, the consensus is now that patients with NPHS2 mutations are at reduced risk for recurrent FSGS following renal transplant [18, 22]. The most common polymorphism is R229Q, with a heterozygote frequency in the general population ranging from 0.03 to 0.13 [23]. R229Q heterozygosity has been associated with microalbuminuria in a Brazilian population [24] and a modest increased risk for FSGS in individuals of European ancestry, but not individuals of African ancestry [23].

CD2-associated protein (CD2AP)

CD2-associated protein was discovered as a binding ligand to the adhesion molecule CD2 during cell–cell interaction of lymphocyte T with antigen presenting cell. This interaction initiates the process of protein segregation, CD2 clustering, and cytoskeletal polarization. The CD2AP gene encodes a protein of 639 amino acids with a deduced molecular mass of approximately 70 kDa, and messenger ribonucleic acid (mRNA) is ubiquitously expressed in human tissues. Cd2ap knock-out mice manifest compromised immune function and they developed proteinuria by 2 weeks of age and died from renal failure at 6–7 weeks of age [25]. In the kidney, the lesion resembles FSGS, with mesangial cell hyperplasia and extracellular matrix deposition. Mice with Cd2ap haploinsufficiency (heterozygous deletion) develop glomerular changes at 9 months of age. They have increased susceptibility to glomerular injury by immune complexes, with likely impairment of the intracellular degradation pathways [26]. CD2AP is the human ortholog of mouse Cd2ap, with 86% identity at the amino acid level. CD2AP (originally named CMS) was identified in the yeast two-hybrid system as interacting partner of p130 (Cas), a cell cycle regulatory protein [27]. CD2AP is a multifunctional adapter molecule, which is localized to the cytoplasm, membrane ruffles, and leading edges of cells. It functions as scaffolding protein involved in the dynamic regulation of the actin cytoskeleton [27]. Two African Americans with primary idiopathic FSGS were found to have a mutation of CD2AP splice acceptor of exon 7 on one allele. This mutation was not seen in control subjects. Although these data are suggestive, family studies showing a consistent relationship between mutation and renal phenotype will be required to establish a pathogenic role. Data supporting a role for CD2AP haploinsufficiency comes from bigenic mouse models of FSGS involving pairwise interaction of CD2AP, Fyn, and synaptopodin [28].

Alpha-actinin-4 (ACTN4)

ACTN4 mutations have been found to be the cause of autosomal dominant FSGS in five families [29, 30]. These patients tend to present in the teenage years or later and progress to end-stage renal disease (ESRD) slowly. The penetrance is variable, and some patients with mutations have a very mild phenotype. Alpha-actinin-4 is an actin-bundling protein associated with cytoskeleton, cell motility, and cancer invasion [31]. It is highly expressed in podocytes, and interacts with synaptopodin and with the tight junction protein membrane-associated guanylate kinase (MAGI)-1 in rat kidney epithelial cells [32]. Mutant alpha-actinin-4 binds actin more tightly that does wild-type alpha-actinin-4, indicating a gain of function mutation. Actn4-null mice develop proteinuria, progressive glomerular disease, and die by several months of age. Histological abnormalities are limited to the kidneys and include diffuse podocyte foot process effacement and globally disrupted morphology [33]. Cultured podocytes from these mice adhere poorly to basement membrane components, suggesting that podocyte loss may contribute to podocytopenia and FSGS [34].

Transient receptor potential cation channel (TRPC6)

TRPC6 belongs to a protein family whose members function as ion channels, mediating capacitative calcium entry into the cell. TRPC6 is a nonselective calcium channel that is activated by diacylglycerol in a membrane-delimited fashion, independently of protein kinase C [35]. TRPC6 is responsible for calcium entry during cell proliferation and is expressed primarily in the placenta, lung, spleen, ovary, and small intestine. In the kidney, TRPC6 is expressed in tubules and glomeruli, including podoctyes and glomerular endothelial cells.
TRPC6 mutations have been identified in six families of European and African geographic ancestry with autosomal dominant FSGS [36, 37]. Each family carried a distinct missense mutation. Age at renal disease presentation ranged from 17 to 52 years, and the duration from onset to ESRD averaged approximately 10 years. TRPC6 interacts with nephrin and podocin, thus localizing the protein to the slit diaphragm complex. Two mutant proteins are associated with increased calcium amplitudes, indicating an activating mutation. The discovery that abnormal TRPC6 disturbs podocyte function suggests that calcium signaling may play a critical role in facilitating podocyte regulation of intracellular function, including control of cytoskeletal and foot process architecture.

Phospholipase C epsilon 1 (PLCE1)

Recently, seven families with infantile or early onset proteinuric renal disease were found to have mutations in PLCE1, which encodes an isoform of phospholipase C, an enzyme that participates in intracellular signaling [4]. In affected individuals, proteinuria was detected between age 2 months and 9 years; renal histology included diffuse mesangial sclerosis and FSGS. Response to treatment included steroid resistance and complete remission to either steroids or cyclosporine. This is the first time a genetic podoctye disease has been associated with steroid-sensitive nephrotic syndrome [4].

Syndromic FSGS

The above genes have mutations associated with FSGS in the absence of extrarenal manifestations. To date, at least five genes have been associated with syndromes of which FSGS is often a part.

WT1

The product of the Wilm’s tumor gene (WT1) is required for normal development of the genitourinary system and mesothelial tissues and is overexpressed in leukemia and various types of solid tumors. WT1 mutations have been identified in patients with Wilm’s tumor, WAGR syndrome (Wilm’s tumor, aniridia, genitourinary abnormalities, and retardation-hypospadias and bilateral cryptorchidism may be seen), Denys-Drash syndrome (urogenital abnormalities, renal failure, pseudohermaphroditism, and Wilm’s tumor), Frasier syndrome (male pseudohermaphroditism and progressive glomerulopathy), and isolated diffuse mesangial sclerosis. WT1 has four major RNA splice variants, and the interactions between the four polypeptide products may play a role in the control of cellular proliferation and differentiation. WT1 acts as either a transcriptional activator or repressor, depending on chromosomal and cellular context. WT1 is expressed predominantly in the kidney and certain hematopoietic cells [38].
The Wilm’s tumor upstream neighbor (WIT1) is located upstream of WT1, localized to chromosome 11p13. WIT1 is transcribed in the opposite direction of WT1, and some WIT1 splice variants include antisense portions of WT1. WIT1 transcripts may play a role in transcriptional regulation of WT1 [39]. WIT1 protein contains 92 amino acids and is expressed in fetal kidney and spleen [40]. Single nucleotide polymorphisms (SNPs) in both WT1 and WIT1 and their common promoter (rs6508, rs2301254, and rs1799937) were significantly associated with HIV-seronegative FSGS, suggesting that variants in these genes may mediate pathogenesis by altering WT1 function [41].

LMX1B

The nail–patella syndrome includes various combinations of dystrophic nails, absent or malformed patellas, elbow contractures, glaucoma, and FSGS [42]. The gene responsible, LMX1B, is a transcription factor required for expression of CD2AP and NPHS2, among other genes [43].

tRNALeu

Mutations in the mitochondrial gene encoding tRNALeu are associated with the MELAS syndrome (myopathy, encephalopathy, lactic acidosis, and stroke-like episodes), typically presenting in infancy or early childhood. There is a maternal inheritance pattern, and the most common mutation is A324G [44, 45]. Patients with this mutation may also present later in childhood or in adulthood with FSGS associated with deafness or diabetes mellitus, or occasionally with sporadic FSGS.

COQ2

Another mitochondriopathy associated with FSGS has been described recently. A mutation in COQ2, encoding an enzyme required for synthesis of coenzyme Q10 (ubiquinone), which is present in all membranes. A COQ2 mutation was found in an infant who presented at 12 months of age with psychomotor delay, optic atrophy, and FSGS [46].

ITGB4

An infant with epidermolysis bullosa, pyloric atresia, and FSGS was found to have a homozygous missense mutation (R1281W) in β4 integrin. The α6β4 integrin binds laminin-5 (a trimer composed of the laminin α5, β3, and γ3 chains). Although the major podocyte integrin is α3β1, the authors showed that podocytes do express low levels of α6β4. Whereas other families with α6β4 integrin mutations and epidermolysis bullosa have been reported, to date, only the one case of FSGS has been reported [47].

Summary

The recent advances in understanding the pathophysiology of FSGS and molecular function of glomerular filtration barrier come in large part from genetic linkage and positional cloning studies. Because a substantial percentage (up to 30%) of sporadic, steroid-resistant nephrotic syndrome patients carry the specific genetic mutation causally linked to FSGS, it may be advisable that all patients who have failed steroids should be offered genetic screening for mutations at the time of or prior to starting more aggressive treatments, as those patients have been shown to unfavorably respond to immunosuppressant therapy [18]. Genetic testing could be performed in several research laboratories and now is also being offered by a commercial laboratory. Genetic testing for familial forms of FSGS might also be clinically informative in situations where living-related donor transplant is considered.
The exact role and function of the newly discovered genes and proteins are being investigated by in vitro and in vivo mechanistic studies. Those genes and proteins interactions seem to change susceptibility to kidney disease progression. Better understanding of their exact role in the development of FSGS may influence future therapies and outcomes in this complex disease.

Questions

(Answers appear following the reference list)
1.
Familial form of FSGS is most commonly associated with mutation of:
a)
NPHS1 (gene encoding nephrin)
 
b)
WT1 (gene encoding Wilm’s tumor protein)
 
c)
CD2AP (gene encoding CD2-associated protein)
 
d)
NPHS2 (gene encoding podocin)
 
 
2.
Mutation of which gene is likely associated with steroid-sensitive nephrotic syndrome:
a)
NPHS1
 
b)
NPHS2
 
c)
TRPC6
 
d)
None of the above
 
 
3.
Although established as a cause and model of FSGS in animals, mutation of which gene is only rarely found in humans:
a)
NPHS1
 
b)
NPHS2
 
c)
CD2AP
 
d)
TRPC6
 
 
4.
Proteins that interact with each other in the vicinity of the slit diaphragm are:
a)
Nephrin, WT1, TRPC6
 
b)
Nephrin, Alpha-actinin-4, Podocin
 
c)
Podocin, CD2AP, Nephrin
 
d)
Nephrin, CD2AP, Alpha-actinin-4
 
 
5.
FSGS that is associated with hypospadias and bilateral cryptorchidism may result from the mutation of:
a)
NPHS2
 
b)
WT1
 
c)
CD2AP
 
d)
TRPC6
 
 

Answers

1:
D
 
2:
D
 
3:
C
 
4:
C
 
5:
B
 
This 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/.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Pädiatrie

Kombi-Abonnement

Mit e.Med Pädiatrie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Pädiatrie, den Premium-Inhalten der pädiatrischen Fachzeitschriften, inklusive einer gedruckten Pädiatrie-Zeitschrift Ihrer Wahl.

e.Med Urologie

Kombi-Abonnement

Mit e.Med Urologie erhalten Sie Zugang zu den urologischen CME-Fortbildungen und Premium-Inhalten der urologischen Fachzeitschriften.

Literatur
1.
Zurück zum Zitat Churg J, Habib R, White RHR (1970) The pathology of the nephrotic syndrome in children. A report of the International Study of Kidney Disease in Children. Lancet 1:1299–1302CrossRef Churg J, Habib R, White RHR (1970) The pathology of the nephrotic syndrome in children. A report of the International Study of Kidney Disease in Children. Lancet 1:1299–1302CrossRef
2.
3.
Zurück zum Zitat Caulfield JP, Farquhar MG (1974) The permeability of glomerular capillaries to graded dextrans. Identification of the basement membrane as the primary filtration barrier. J Cell Biol 63:883–903PubMedPubMedCentralCrossRef Caulfield JP, Farquhar MG (1974) The permeability of glomerular capillaries to graded dextrans. Identification of the basement membrane as the primary filtration barrier. J Cell Biol 63:883–903PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Hinkes B, Wiggins RC, Gbadegesin R, Vlangos CN, Seelow D, Nurnberg G, Garg P, Verma R, Chaib H, Hoskins BE, Ashraf S, Becker C, Hennies HC, Goyal M, Wharram BL, Schachter AD, Mudumana S, Drummond I, Kerjaschki D, Waldherr R, Dietrich A, Ozaltin F, Bakkaloglu A, Cleper R, Basel-Vanagaite L, Pohl M, Griebel M, Tsygin AN, Soylu A, Muller D, Sorli CS, Bunney TD, Katan M, Liu J, Attanasio M, O’toole JF, Hasselbacher K, Mucha B, Otto EA, Airik R, Kispert A, Kelley GG, Smrcka AV, Gudermann T, Holzman LB, Nurnberg P, Hildebrandt F (2006) Positional cloning uncovers mutations in PLCE1 responsible for a nephrotic syndrome variant that may be reversible. Nat Genet 38:1397–1405PubMedCrossRef Hinkes B, Wiggins RC, Gbadegesin R, Vlangos CN, Seelow D, Nurnberg G, Garg P, Verma R, Chaib H, Hoskins BE, Ashraf S, Becker C, Hennies HC, Goyal M, Wharram BL, Schachter AD, Mudumana S, Drummond I, Kerjaschki D, Waldherr R, Dietrich A, Ozaltin F, Bakkaloglu A, Cleper R, Basel-Vanagaite L, Pohl M, Griebel M, Tsygin AN, Soylu A, Muller D, Sorli CS, Bunney TD, Katan M, Liu J, Attanasio M, O’toole JF, Hasselbacher K, Mucha B, Otto EA, Airik R, Kispert A, Kelley GG, Smrcka AV, Gudermann T, Holzman LB, Nurnberg P, Hildebrandt F (2006) Positional cloning uncovers mutations in PLCE1 responsible for a nephrotic syndrome variant that may be reversible. Nat Genet 38:1397–1405PubMedCrossRef
5.
Zurück zum Zitat Kestila M, Lenkkeri U, Mannikko M, Lamerdin J, McCready P, Putaala H, Ruotsalainen V, Morita T, Nissinen M, Herva R, Kashtan CE, Peltonen L, Holmberg C, Olsen A, Tryggvason K (1998) Positionally cloned gene for a novel glomerular protein–nephrin–is mutated in congenital nephrotic syndrome. Mol Cell 1:575–582PubMedCrossRef Kestila M, Lenkkeri U, Mannikko M, Lamerdin J, McCready P, Putaala H, Ruotsalainen V, Morita T, Nissinen M, Herva R, Kashtan CE, Peltonen L, Holmberg C, Olsen A, Tryggvason K (1998) Positionally cloned gene for a novel glomerular protein–nephrin–is mutated in congenital nephrotic syndrome. Mol Cell 1:575–582PubMedCrossRef
6.
Zurück zum Zitat Koziell A, Grech V, Hussain S, Lee G, Lenkkeri U, Tryggvason K, Scambler P (2002) Genotype/phenotype correlations of NPHS1 and NPHS2 mutations in nephrotic syndrome advocate a functional inter-relationship in glomerular filtration. Hum Mol Genet 11:379–388PubMedCrossRef Koziell A, Grech V, Hussain S, Lee G, Lenkkeri U, Tryggvason K, Scambler P (2002) Genotype/phenotype correlations of NPHS1 and NPHS2 mutations in nephrotic syndrome advocate a functional inter-relationship in glomerular filtration. Hum Mol Genet 11:379–388PubMedCrossRef
7.
Zurück zum Zitat Wartiovaara J, Ofverstedt LG, Khoshnoodi J, Zhang J, Makela E, Sandin S, Ruotsalainen V, Cheng RH, Jalanko H, Skoglund U, Tryggvason K (2004) Nephrin strands contribute to a porous slit diaphragm scaffold as revealed by electron tomography. J Clin Invest 114:1475–1483PubMedPubMedCentralCrossRef Wartiovaara J, Ofverstedt LG, Khoshnoodi J, Zhang J, Makela E, Sandin S, Ruotsalainen V, Cheng RH, Jalanko H, Skoglund U, Tryggvason K (2004) Nephrin strands contribute to a porous slit diaphragm scaffold as revealed by electron tomography. J Clin Invest 114:1475–1483PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Jones N, Blasutig IM, Eremina V, Ruston JM, Bladt F, Li H, Huang H, Larose L, Li SSC, Takano T, Quaggin, SE, Pawson T (2006) Nck adaptor proteins link nephrin to the actin cytoskeleton of kidney podocytes. Nature 440:818–823CrossRefPubMed Jones N, Blasutig IM, Eremina V, Ruston JM, Bladt F, Li H, Huang H, Larose L, Li SSC, Takano T, Quaggin, SE, Pawson T (2006) Nck adaptor proteins link nephrin to the actin cytoskeleton of kidney podocytes. Nature 440:818–823CrossRefPubMed
9.
Zurück zum Zitat Beltcheva O, Martin P, Lenkkeri U, Tryggvason K (2001) Mutation spectrum in the nephrin gene (NPHS1) in congenital nephrotic syndrome. Hum Mutat 17:368–373PubMedCrossRef Beltcheva O, Martin P, Lenkkeri U, Tryggvason K (2001) Mutation spectrum in the nephrin gene (NPHS1) in congenital nephrotic syndrome. Hum Mutat 17:368–373PubMedCrossRef
10.
Zurück zum Zitat Bolk S, Puffenberger EG, Hudson J, Morton DH, Chakravarti A (1999) Elevated frequency and allelic heterogeneity of congenital nephrotic syndrome, Finnish type, in the Old Order Mennonites. Am J Hum Genet 65:1785–1790PubMedPubMedCentralCrossRef Bolk S, Puffenberger EG, Hudson J, Morton DH, Chakravarti A (1999) Elevated frequency and allelic heterogeneity of congenital nephrotic syndrome, Finnish type, in the Old Order Mennonites. Am J Hum Genet 65:1785–1790PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Aya K, Tanaka H, Seino Y (2000) Novel mutation in the nephrin gene of a Japanese patient with congenital nephrotic syndrome of the Finnish type. Kidney Int 57:401–404PubMedCrossRef Aya K, Tanaka H, Seino Y (2000) Novel mutation in the nephrin gene of a Japanese patient with congenital nephrotic syndrome of the Finnish type. Kidney Int 57:401–404PubMedCrossRef
12.
Zurück zum Zitat Boute N, Gribouval O, Roselli S, Benessy F, Lee H, Fuchshuber A, Dahan K, Gubler MC, Niaudet P, Antignac C (2000) NPHS2, encoding the glomerular protein podocin, is mutated in autosomal recessive steroid-resistant nephrotic syndrome. Nat Genet 24:349–354CrossRefPubMed Boute N, Gribouval O, Roselli S, Benessy F, Lee H, Fuchshuber A, Dahan K, Gubler MC, Niaudet P, Antignac C (2000) NPHS2, encoding the glomerular protein podocin, is mutated in autosomal recessive steroid-resistant nephrotic syndrome. Nat Genet 24:349–354CrossRefPubMed
13.
Zurück zum Zitat Schwarz K, Simons M, Reiser J, Saleem MA, Faul C, Kriz W, Shaw AS, Holzman LB, Mundel P (2001) Podocin, a raft-associated component of the glomerular slit diaphragm, interacts with CD2AP and nephrin. J Clin Invest 108:1621–1629PubMedPubMedCentralCrossRef Schwarz K, Simons M, Reiser J, Saleem MA, Faul C, Kriz W, Shaw AS, Holzman LB, Mundel P (2001) Podocin, a raft-associated component of the glomerular slit diaphragm, interacts with CD2AP and nephrin. J Clin Invest 108:1621–1629PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Huber TB, Simons M, Hartleben B, Sernetz L, Schmidts M, Gundlach E, Saleem MA, Walz G, Benzing T (2003) Molecular basis of the functional podocin-nephrin complex: mutations in the NPHS2 gene disrupt nephrin targeting to lipid raft microdomains. Hum Mol Genet 12:3397–3405CrossRefPubMed Huber TB, Simons M, Hartleben B, Sernetz L, Schmidts M, Gundlach E, Saleem MA, Walz G, Benzing T (2003) Molecular basis of the functional podocin-nephrin complex: mutations in the NPHS2 gene disrupt nephrin targeting to lipid raft microdomains. Hum Mol Genet 12:3397–3405CrossRefPubMed
15.
Zurück zum Zitat Roselli S, Heidet L, Sich M, Henger A, Kretzler M, Gubler MC, Antignac C (2004) Early glomerular filtration defect and severe renal disease in podocin-deficient mice. Mol Cell Biol 24:550–560PubMedPubMedCentralCrossRef Roselli S, Heidet L, Sich M, Henger A, Kretzler M, Gubler MC, Antignac C (2004) Early glomerular filtration defect and severe renal disease in podocin-deficient mice. Mol Cell Biol 24:550–560PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Caridi G, Bertelli R, Carrea A, Di Duca M, Catarsi P, Artero M, Carraro M, Zennaro C, Candiano G, Musante L, Seri M, Ginevri F, Perfumo F, Ghiggeri GM (2001) Prevalence, genetics, and clinical features of patients carrying podocin mutations in steroid-resistant nonfamilial focal segmental glomerulosclerosis. J Am Soc Nephrol 12:2742–2746PubMedCrossRef Caridi G, Bertelli R, Carrea A, Di Duca M, Catarsi P, Artero M, Carraro M, Zennaro C, Candiano G, Musante L, Seri M, Ginevri F, Perfumo F, Ghiggeri GM (2001) Prevalence, genetics, and clinical features of patients carrying podocin mutations in steroid-resistant nonfamilial focal segmental glomerulosclerosis. J Am Soc Nephrol 12:2742–2746PubMedCrossRef
17.
Zurück zum Zitat Tsukaguchi H, Sudhakar A, Le TC, Nguyen T, Yao J, Schwimmer JA, Schachter AD, Poch E, Abreu PF, Appel GB, Pereira AB, Kalluri R, Pollak MR (2002) NPHS2 mutations in late-onset focal segmental glomerulosclerosis: R229Q is a common disease-associated allele. J Clin Invest 110:1659–1666PubMedPubMedCentralCrossRef Tsukaguchi H, Sudhakar A, Le TC, Nguyen T, Yao J, Schwimmer JA, Schachter AD, Poch E, Abreu PF, Appel GB, Pereira AB, Kalluri R, Pollak MR (2002) NPHS2 mutations in late-onset focal segmental glomerulosclerosis: R229Q is a common disease-associated allele. J Clin Invest 110:1659–1666PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Ruf RG, Lichtenberger A, Karle SM, Haas JP, Anacleto FE, Schultheiss M, Zalewski I, Imm A, Ruf EM, Mucha B, Bagga A, Neuhaus T, Fuchshuber A, Bakkaloglu A, Hildebrandt F, Arbeitsgemeinschaft Fur Padiatrische Nephrologie Study Group (2004) Patients with mutations in NPHS2 (podocin) do not respond to standard steroid treatment of nephrotic syndrome. J Am Soc Nephrol 15:722–732CrossRefPubMed Ruf RG, Lichtenberger A, Karle SM, Haas JP, Anacleto FE, Schultheiss M, Zalewski I, Imm A, Ruf EM, Mucha B, Bagga A, Neuhaus T, Fuchshuber A, Bakkaloglu A, Hildebrandt F, Arbeitsgemeinschaft Fur Padiatrische Nephrologie Study Group (2004) Patients with mutations in NPHS2 (podocin) do not respond to standard steroid treatment of nephrotic syndrome. J Am Soc Nephrol 15:722–732CrossRefPubMed
19.
Zurück zum Zitat Aucella F, De Bonis P, Gatta G, Muscarella LA, Vigilante M, di Giorgio G, D’Errico M, Zelante L, Stallone C, Bisceglia L (2005) Molecular analysis of NPHS2 and ACTN4 genes in a series of 33 Italian patients affected by adult-onset nonfamilial focal segmental glomerulosclerosis. Nephron Clin Pract 99:c31–c36PubMedCrossRef Aucella F, De Bonis P, Gatta G, Muscarella LA, Vigilante M, di Giorgio G, D’Errico M, Zelante L, Stallone C, Bisceglia L (2005) Molecular analysis of NPHS2 and ACTN4 genes in a series of 33 Italian patients affected by adult-onset nonfamilial focal segmental glomerulosclerosis. Nephron Clin Pract 99:c31–c36PubMedCrossRef
20.
Zurück zum Zitat Monteiro EJ, Pereira AC, Pereira AB, Krieger JE, Mastroianni-Kirsztajn G (2006) NPHS2 mutations in adult patients with primary focal segmental glomerulosclerosis. J Nephrol 19:366–371PubMed Monteiro EJ, Pereira AC, Pereira AB, Krieger JE, Mastroianni-Kirsztajn G (2006) NPHS2 mutations in adult patients with primary focal segmental glomerulosclerosis. J Nephrol 19:366–371PubMed
21.
Zurück zum Zitat Maruyama K, Iijima K, Ikeda M, Kitamura A, Tsukaguchi H, Yoshiya K, Hoshii S, Wada N, Uemura O, Satomura K, Honda M, Yoshikawa N (2003) NPHS2 mutations in sporadic steroid-resistant nephrotic syndrome in Japanese children. Pediatr Nephrol 18:412–416CrossRefPubMed Maruyama K, Iijima K, Ikeda M, Kitamura A, Tsukaguchi H, Yoshiya K, Hoshii S, Wada N, Uemura O, Satomura K, Honda M, Yoshikawa N (2003) NPHS2 mutations in sporadic steroid-resistant nephrotic syndrome in Japanese children. Pediatr Nephrol 18:412–416CrossRefPubMed
22.
Zurück zum Zitat Weber S, Gribouval O, Esquivel EL, Moriniere V, Tete MJ, Legendre C, Niaudet P, Antignac C (2004) NPHS2 mutation analysis shows genetic heterogeneity of steroid-resistant nephrotic syndrome and low post-transplant recurrence. Kidney Int 66:571–579CrossRefPubMed Weber S, Gribouval O, Esquivel EL, Moriniere V, Tete MJ, Legendre C, Niaudet P, Antignac C (2004) NPHS2 mutation analysis shows genetic heterogeneity of steroid-resistant nephrotic syndrome and low post-transplant recurrence. Kidney Int 66:571–579CrossRefPubMed
23.
Zurück zum Zitat Franceschini N, North KE, Kopp JB, McKenzie L, Winkler C (2006) NPHS2 gene, nephrotic syndrome and focal segmental glomerulosclerosis: a HuGE review. Genet Med 8:63–75CrossRefPubMed Franceschini N, North KE, Kopp JB, McKenzie L, Winkler C (2006) NPHS2 gene, nephrotic syndrome and focal segmental glomerulosclerosis: a HuGE review. Genet Med 8:63–75CrossRefPubMed
24.
Zurück zum Zitat Pereira AC, Pereira AB, Mota GF, Cunha RS, Herkenhoff FL, Pollak MR, Mill JG, Krieger JE (2004) NPHS2 R229Q functional variant is associated with microalbuminuria in the general population. Kidney Int 65:1026–1030CrossRefPubMed Pereira AC, Pereira AB, Mota GF, Cunha RS, Herkenhoff FL, Pollak MR, Mill JG, Krieger JE (2004) NPHS2 R229Q functional variant is associated with microalbuminuria in the general population. Kidney Int 65:1026–1030CrossRefPubMed
25.
Zurück zum Zitat Shih NY, Li J, Karpitskii V, Nguyen A, Dustin ML, Kanagawa O, Miner JH, Shaw AS (1999) Congenital nephrotic syndrome in mice lacking CD2-associated protein. Science 286:312–315CrossRefPubMed Shih NY, Li J, Karpitskii V, Nguyen A, Dustin ML, Kanagawa O, Miner JH, Shaw AS (1999) Congenital nephrotic syndrome in mice lacking CD2-associated protein. Science 286:312–315CrossRefPubMed
26.
Zurück zum Zitat Kim JM, Wu H, Green G, Winkler CA, Kopp JB, Miner JH, Unanue ER, Shaw AS (2003) CD2-associated protein haploinsufficiency is linked to glomerular disease susceptibility. Science 300:1298–1300CrossRefPubMed Kim JM, Wu H, Green G, Winkler CA, Kopp JB, Miner JH, Unanue ER, Shaw AS (2003) CD2-associated protein haploinsufficiency is linked to glomerular disease susceptibility. Science 300:1298–1300CrossRefPubMed
27.
Zurück zum Zitat Kirsch KH, Georgescu MM, Ishimaru S, Hanafusa H (1999) CMS: an adapter molecule involved in cytoskeletal rearrangements. Proc Nat Acad Sci USA 96:6211–6216PubMedCrossRefPubMedCentral Kirsch KH, Georgescu MM, Ishimaru S, Hanafusa H (1999) CMS: an adapter molecule involved in cytoskeletal rearrangements. Proc Nat Acad Sci USA 96:6211–6216PubMedCrossRefPubMedCentral
28.
Zurück zum Zitat Huber TB, Kwoh C, Wu H, Asanuma K, Godel M, Hartleben B, Blumer KJ, Miner JH, Mundel P, Shaw AS (2006) Bigenic mouse models of focal segmental glomerulosclerosis involving pairwise interaction of CD2AP, Fyn, and synaptopodin. J Clin Invest 116:1337–1345PubMedPubMedCentralCrossRef Huber TB, Kwoh C, Wu H, Asanuma K, Godel M, Hartleben B, Blumer KJ, Miner JH, Mundel P, Shaw AS (2006) Bigenic mouse models of focal segmental glomerulosclerosis involving pairwise interaction of CD2AP, Fyn, and synaptopodin. J Clin Invest 116:1337–1345PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Kaplan JM, Kim SH, North KN, Rennke H, Correia LA, Tong H-Q, Mathis BJ, Rodriguez-Perez J-C, Allen PG, Beggs AH, Pollak MR (2000) Mutations in ACTN4, encoding alpha-actinin-4, cause familial focal segmental glomerulosclerosis. Nat Genet 24:251–256PubMedCrossRef Kaplan JM, Kim SH, North KN, Rennke H, Correia LA, Tong H-Q, Mathis BJ, Rodriguez-Perez J-C, Allen PG, Beggs AH, Pollak MR (2000) Mutations in ACTN4, encoding alpha-actinin-4, cause familial focal segmental glomerulosclerosis. Nat Genet 24:251–256PubMedCrossRef
30.
Zurück zum Zitat Weins A, Kenlan P, Herbert S, Le TC, Villegas I, Kaplan BS, Appel GB, Pollak MR (2005) Mutational and Biological Analysis of alpha-actinin-4 in focal segmental glomerulosclerosis. J Am Soc Nephrol 16:3694–3701PubMedCrossRef Weins A, Kenlan P, Herbert S, Le TC, Villegas I, Kaplan BS, Appel GB, Pollak MR (2005) Mutational and Biological Analysis of alpha-actinin-4 in focal segmental glomerulosclerosis. J Am Soc Nephrol 16:3694–3701PubMedCrossRef
31.
Zurück zum Zitat Honda K, Yamada T, Endo R, Ino Y, Gotoh M, Tsuda H, Yamada Y, Chiba H, Hirohashi S (1998) Actinin-4, a novel actin-bundling protein associated with cell motility and cancer invasion. J Cell Biol 140:1383–1393PubMedPubMedCentralCrossRef Honda K, Yamada T, Endo R, Ino Y, Gotoh M, Tsuda H, Yamada Y, Chiba H, Hirohashi S (1998) Actinin-4, a novel actin-bundling protein associated with cell motility and cancer invasion. J Cell Biol 140:1383–1393PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Patrie KM, Drescher AJ, Welihinda A, Mundel P, Margolis B (2002) Interaction of two actin-binding proteins, synaptopodin and alpha-actinin-4, with the tight junction protein MAGI-1. J Biol Chem 277:30183–30190PubMedCrossRef Patrie KM, Drescher AJ, Welihinda A, Mundel P, Margolis B (2002) Interaction of two actin-binding proteins, synaptopodin and alpha-actinin-4, with the tight junction protein MAGI-1. J Biol Chem 277:30183–30190PubMedCrossRef
33.
Zurück zum Zitat Kos CH, Le TC, Sinha S, Henderson JM, Kim SH, Sugimoto H, Kalluri R, Gerszten RE, Pollak MR (2003) Mice deficient in alpha-actinin-4 have severe glomerular disease. J Clin Invest 111:1683–1690PubMedPubMedCentralCrossRef Kos CH, Le TC, Sinha S, Henderson JM, Kim SH, Sugimoto H, Kalluri R, Gerszten RE, Pollak MR (2003) Mice deficient in alpha-actinin-4 have severe glomerular disease. J Clin Invest 111:1683–1690PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Dandapani SV, Sugimoto H, Matthews BD, Kolb RJ, Sinha S, Gerszten RE, Zhou J, Ingber DE, Kalluri R, Pollak MR (2007) Alpha -actinin-4 is required for normal podocyte adhesion. J Biol Chem 282:467–477PubMedCrossRef Dandapani SV, Sugimoto H, Matthews BD, Kolb RJ, Sinha S, Gerszten RE, Zhou J, Ingber DE, Kalluri R, Pollak MR (2007) Alpha -actinin-4 is required for normal podocyte adhesion. J Biol Chem 282:467–477PubMedCrossRef
35.
Zurück zum Zitat Hofmann T, Obukhov AG, Schaefer M, Harteneck C, Gudermann T, Schultz G (1999) Direct activation of human TRPC6 and TRPC3 channels by diacylglycerol. Nature 397:259–263PubMedCrossRef Hofmann T, Obukhov AG, Schaefer M, Harteneck C, Gudermann T, Schultz G (1999) Direct activation of human TRPC6 and TRPC3 channels by diacylglycerol. Nature 397:259–263PubMedCrossRef
36.
Zurück zum Zitat Reiser J, Polu KR, Moller CC, Kenlan P, Altintas MM, Wei C, Faul C, Herbert S, Villegas I, Avila-Casado C, McGee M, Sugimoto H, Brown D, Kalluri R, Mundel P, Smith PL, Clapham DE, Pollak MR (2005) TRPC6 is a glomerular slit diaphragm-associated channel required for normal renal function. Nat Genet 37:739–744PubMedPubMedCentralCrossRef Reiser J, Polu KR, Moller CC, Kenlan P, Altintas MM, Wei C, Faul C, Herbert S, Villegas I, Avila-Casado C, McGee M, Sugimoto H, Brown D, Kalluri R, Mundel P, Smith PL, Clapham DE, Pollak MR (2005) TRPC6 is a glomerular slit diaphragm-associated channel required for normal renal function. Nat Genet 37:739–744PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Winn MP, Conlon PJ, Lynn KL, Farrington MK, Creazzo T, Hawkins AF, Daskalakis N, Kwan SY, Ebersviller S, Burchette JL, Pericak-Vance MA, Howell DN, Vance JM, Rosenberg PB (2005) A mutation in the TRPC6 cation channel causes familial focal segmental glomerulosclerosis. Science 308:1801–1804CrossRefPubMed Winn MP, Conlon PJ, Lynn KL, Farrington MK, Creazzo T, Hawkins AF, Daskalakis N, Kwan SY, Ebersviller S, Burchette JL, Pericak-Vance MA, Howell DN, Vance JM, Rosenberg PB (2005) A mutation in the TRPC6 cation channel causes familial focal segmental glomerulosclerosis. Science 308:1801–1804CrossRefPubMed
38.
Zurück zum Zitat Gessler M, Poustka A, Cavenee W, Neve RL, Orkin SH, Bruns GAP (1990) Homozygous deletion in Wilms tumours of a zinc-finger gene identified by chromosome jumping. Nature 343:774–778PubMedCrossRef Gessler M, Poustka A, Cavenee W, Neve RL, Orkin SH, Bruns GAP (1990) Homozygous deletion in Wilms tumours of a zinc-finger gene identified by chromosome jumping. Nature 343:774–778PubMedCrossRef
39.
Zurück zum Zitat Campbell CE, Huang A, Gurney AL, Kessler PM, Hewitt JA, Williams BRG (1994) Antisense transcripts and protein binding motifs within the Wilms tumour (WT1) locus. Oncogene 9:583–595PubMed Campbell CE, Huang A, Gurney AL, Kessler PM, Hewitt JA, Williams BRG (1994) Antisense transcripts and protein binding motifs within the Wilms tumour (WT1) locus. Oncogene 9:583–595PubMed
40.
Zurück zum Zitat Huang A, Campbell CE, Bonetta L, McAndrews-Hill MS, Chilton-MacNeill S, Coppes MJ, Law DJ, Feinberg AP, Yeger H, Williams BRG (1990) Tissue, developmental, and tumor-specific expression of divergent transcripts in Wilms tumor. Science 250:991–994PubMedCrossRef Huang A, Campbell CE, Bonetta L, McAndrews-Hill MS, Chilton-MacNeill S, Coppes MJ, Law DJ, Feinberg AP, Yeger H, Williams BRG (1990) Tissue, developmental, and tumor-specific expression of divergent transcripts in Wilms tumor. Science 250:991–994PubMedCrossRef
41.
Zurück zum Zitat Orloff MS, Iyengar SK, Winkler CA, Goddard KA, Dart RA, Ahuja TS, Mokrzycki M, Briggs WA, Korbet SM, Kimmel PL, Simon EE, Trachtman H, Vlahov D, Michel DM, Berns JS, Smith MC, Schelling JR, Sedor JR, Kopp JB (2005) Variants in the Wilms’ tumor gene are associated with focal segmental glomerulosclerosis in the African American population. Physiol Genomics 21:212–221CrossRefPubMed Orloff MS, Iyengar SK, Winkler CA, Goddard KA, Dart RA, Ahuja TS, Mokrzycki M, Briggs WA, Korbet SM, Kimmel PL, Simon EE, Trachtman H, Vlahov D, Michel DM, Berns JS, Smith MC, Schelling JR, Sedor JR, Kopp JB (2005) Variants in the Wilms’ tumor gene are associated with focal segmental glomerulosclerosis in the African American population. Physiol Genomics 21:212–221CrossRefPubMed
42.
Zurück zum Zitat Bongers EM, Gubler MC, Knoers NV (2002) Nail-patella syndrome. Overview on clinical and molecular findings. Pediatr Nephrol 17:703–712PubMedCrossRef Bongers EM, Gubler MC, Knoers NV (2002) Nail-patella syndrome. Overview on clinical and molecular findings. Pediatr Nephrol 17:703–712PubMedCrossRef
43.
Zurück zum Zitat Miner JH, Morello R, Andrews KL, Li C, Antignac C, Shaw AS, Lee B (2002) Transcriptional induction of slit diaphragm genes by Lmx1b is required in podocyte differentiation. J Clin Invest 109:1065–1072PubMedPubMedCentralCrossRef Miner JH, Morello R, Andrews KL, Li C, Antignac C, Shaw AS, Lee B (2002) Transcriptional induction of slit diaphragm genes by Lmx1b is required in podocyte differentiation. J Clin Invest 109:1065–1072PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Hotta O, Inoue CN, Miyabayashi S, Furuta T, Takeuchi A, Taguma Y (2001) Clinical and pathologic features of focal segmental glomerulosclerosis with mitochondrial tRNALeu(UUR) gene mutation. Kidney Int 59:1236–1243CrossRefPubMed Hotta O, Inoue CN, Miyabayashi S, Furuta T, Takeuchi A, Taguma Y (2001) Clinical and pathologic features of focal segmental glomerulosclerosis with mitochondrial tRNALeu(UUR) gene mutation. Kidney Int 59:1236–1243CrossRefPubMed
45.
Zurück zum Zitat Guery B, Choukroun G, Noel LH, Clavel P, Rotig A, Lebon S, Rustin P, Bellane-Chantelot C, Mougenot B, Grunfeld JP, Chauveau D (2003) The spectrum of systemic involvement in adults presenting with renal lesion and mitochondrial tRNA(Leu) gene mutation. J Am Soc Nephrol 14:2099–2108CrossRefPubMed Guery B, Choukroun G, Noel LH, Clavel P, Rotig A, Lebon S, Rustin P, Bellane-Chantelot C, Mougenot B, Grunfeld JP, Chauveau D (2003) The spectrum of systemic involvement in adults presenting with renal lesion and mitochondrial tRNA(Leu) gene mutation. J Am Soc Nephrol 14:2099–2108CrossRefPubMed
46.
Zurück zum Zitat Quinzii C, Naini A, Salviati L, Trevisson E, Navas P, Dimauro S, Hirano M (2006) A Mutation in Para-Hydroxybenzoate-Polyprenyl Transferase (COQ2) Causes Primary Coenzyme Q10 Deficiency. Am J Hum Genet 78:345–349PubMedCrossRef Quinzii C, Naini A, Salviati L, Trevisson E, Navas P, Dimauro S, Hirano M (2006) A Mutation in Para-Hydroxybenzoate-Polyprenyl Transferase (COQ2) Causes Primary Coenzyme Q10 Deficiency. Am J Hum Genet 78:345–349PubMedCrossRef
47.
Zurück zum Zitat Pulkkinen L, Rouan F, Bruckner-Tuderman L, Wallerstein R, Garzon M, Brown T, Smith L, Carter W, Uitto J (1998) Novel ITGB4 mutations in lethal and nonlethal variants of epidermolysis bullosa with pyloric atresia: missense versus nonsense. Am J Hum Genet 63:1376–1387PubMedPubMedCentralCrossRef Pulkkinen L, Rouan F, Bruckner-Tuderman L, Wallerstein R, Garzon M, Brown T, Smith L, Carter W, Uitto J (1998) Novel ITGB4 mutations in lethal and nonlethal variants of epidermolysis bullosa with pyloric atresia: missense versus nonsense. Am J Hum Genet 63:1376–1387PubMedPubMedCentralCrossRef
Metadaten
Titel
Genetics of focal segmental glomerulosclerosis
verfasst von
Robert P. Woroniecki
Jeffrey B. Kopp
Publikationsdatum
01.05.2007
Verlag
Springer Berlin Heidelberg
Erschienen in
Pediatric Nephrology / Ausgabe 5/2007
Print ISSN: 0931-041X
Elektronische ISSN: 1432-198X
DOI
https://doi.org/10.1007/s00467-007-0445-y

Weitere Artikel der Ausgabe 5/2007

Pediatric Nephrology 5/2007 Zur Ausgabe

ADHS-Medikation erhöht das kardiovaskuläre Risiko

16.05.2024 Herzinsuffizienz Nachrichten

Erwachsene, die Medikamente gegen das Aufmerksamkeitsdefizit-Hyperaktivitätssyndrom einnehmen, laufen offenbar erhöhte Gefahr, an Herzschwäche zu erkranken oder einen Schlaganfall zu erleiden. Es scheint eine Dosis-Wirkungs-Beziehung zu bestehen.

Erstmanifestation eines Diabetes-Typ-1 bei Kindern: Ein Notfall!

16.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Manifestiert sich ein Typ-1-Diabetes bei Kindern, ist das ein Notfall – ebenso wie eine diabetische Ketoazidose. Die Grundsäulen der Therapie bestehen aus Rehydratation, Insulin und Kaliumgabe. Insulin ist das Medikament der Wahl zur Behandlung der Ketoazidose.

Frühe Hypertonie erhöht späteres kardiovaskuläres Risiko

Wie wichtig es ist, pädiatrische Patienten auf Bluthochdruck zu screenen, zeigt eine kanadische Studie: Hypertone Druckwerte in Kindheit und Jugend steigern das Risiko für spätere kardiovaskuläre Komplikationen.

Betalaktam-Allergie: praxisnahes Vorgehen beim Delabeling

16.05.2024 Pädiatrische Allergologie Nachrichten

Die große Mehrheit der vermeintlichen Penicillinallergien sind keine. Da das „Etikett“ Betalaktam-Allergie oft schon in der Kindheit erworben wird, kann ein frühzeitiges Delabeling lebenslange Vorteile bringen. Ein Team von Pädiaterinnen und Pädiatern aus Kanada stellt vor, wie sie dabei vorgehen.

Update Pädiatrie

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