Sie sind auf Seite 1von 8

J Nephrol

DOI 10.1007/s40620-016-0315-4

REVIEW

Lessons from genetics: is it time to revise the therapeutic approach


to children with steroid-resistant nephrotic syndrome?
Francesca Becherucci1 Benedetta Mazzinghi1 Aldesia Provenzano2

Luisa Murer3 Sabrina Giglio2,4 Paola Romagnani1,4

Received: 2 February 2016 / Accepted: 29 April 2016


Italian Society of Nephrology 2016

Abstract Primitive nephrotic syndrome is one of the most Keywords Steroid-resistant nephrotic syndrome 
common glomerular diseases in childhood and represents Mutation  Genetics  Children  Gene  Immunosuppressive
the clinical manifestation of various pathologic changes in therapy
the kidney. In children, nephrotic syndrome is classified
based on the initial response to empiric corticosteroid
treatment, which is considered as the best predictor of Introduction
patients final outcome. The advent of next-generation
sequencing technology showed that genetic alterations in Primitive nephrotic syndrome (NS) represents the most
structural genes of the podocyte can be recognized in a common glomerular disease in childhood, with an annual
significant proportion of not only familial or syndromic incidence in unselected cohorts of children of approxi-
patients with steroid-resistant nephrotic syndrome (SRNS), mately 26:100,000 per year in the United States and
but also of sporadic cases, raising the question of whether it Europe [13]. The global prevalence is estimated to be
is time to update current protocols of patient care. In this around 16:100,000 children [2]. Primary NS is defined by
review, we discuss the implications derived from several the absence of recognizable causes of kidney damage that
studies describing a high prevalence in children with SRNS conversely characterize secondary forms (e.g. viral infec-
of pathogenic mutations in a group of genes and their tions, vasculitis, systemic lupus erythematosus). NS is the
unresponsiveness to immunosuppressive therapy. We pro- clinical manifestation of various pathologic changes in the
pose a diagnostic and therapeutic algorithm to reduce the kidney [4], the most common in children being minimal
exposure to immunosuppressants in individuals with change disease (MCD), focal segmental glomerulosclerosis
unresponsive forms of the disease, sparing patients the (FSGS) and diffuse mesangial sclerosis (DMS). Interest-
untoward side effects of prolonged ineffective treatments, ingly enough, numerous studies have demonstrated that the
and at the same time guaranteeing the optimal immuno- clinical picture does not necessarily correlate with patho-
suppressive or other new therapy in potentially responsive logic findings, although FSGS tends to be a more common
patients. finding in patients with steroid-resistant NS (SRNS) [3, 5].
In clinical practice, NS is classified based on patients
& Paola Romagnani initial response to corticosteroid therapy, especially in the
p.romagnani@dfc.unifi.it; romagnani@meyer.it
pediatric population. The great majority of children (ap-
1
Nephrology and Dialysis Unit, Meyer Childrens Hospital, proximately 8090 %) who present with NS respond
Florence, Italy promptly to steroid therapy, which is therefore empirically
2
Medical Genetics Unit, Meyer Childrens Hospital, Florence, performed at the onset of the clinical picture. This group of
Italy patients is defined according to their clinical behavior as
3
Department of Pediatrics, University of Padua, Padua, Italy steroid-sensitive. However, about 1020 % of patients fail
4 to respond to initial steroid treatment and are classified as
Department of Biomedical Experimental and Clinical
Sciences Mario Serio, University of Florence, Viale steroid-resistant [1]. Even though we still lack complete
Pieraccini 6, 50139 Florence, Italy accordance on how to define resistance to steroids [6, 7], it

123
J Nephrol

is commonly accepted that a patient presenting with NS CKD is tracked, even though evidence exists that remission
and failing to achieve remission after 68 weeks of treat- and even regression of glomerular damage can occur [13,
ment with corticosteroids could be defined as steroid-re- 1518].
sistant [6, 8]. SRNS represents the second most frequent In recent years, a vast amount of studies have therefore
cause of chronic kidney disease (CKD) in children after been focused on this issue, even for NS, with genetic
congenital abnormalities of the kidney and the urinary tract alterations of podocytes structure progressively emerging
(CAKUT), and is responsible for a high fraction of cases of as an important mechanism in the pathogenesis of SRNS,
end-stage renal disease (ESRD) in the pediatric population particularly in children.
[9], that usually occurs within 210 years of diagnosis [2,
9, 10]. In addition, children affected by SRNS have a The role of genetics
considerably high risk of infections, thromboembolic
events, growth retardation, acute kidney injury, and CKD Inherited diseases are caused by mutations in genes whose
that significantly reduce life expectancy, as well as of expression is critical for the development, survival and
developing potentially threatening side effects from pro- function of a specific tissue. They are predominantly
longed and often combined immunosuppressive treatments. monogenic disorders, since mutations of a single gene are
The classification of NS by the response to steroids in sufficient to cause the disease. First reports on the possible
steroid-sensitive NS (SSNS) and SRNS highlights the role of genetic abnormalities in determining podocyte
possibility that different pathophysiologic mechanisms dysfunction date back to 1998, when, by means of posi-
confer the response to steroids [3]. Although the patho- tional cloning studies in neonates with congenital NS and
genesis of NS is still not completely understood, con- their families, mutations in the NPHS1 gene encoding for
verging evidence identifies the glomerular filtration barrier the protein nephrin were recognized as the cause of con-
(GFB) as the primary site of different kinds of insults that genital NS of the Finnish type, an autosomal recessive form
finally lead to protein leakage and to the development of of NS that is not responsive to steroid treatment [19].
nephrotic proteinuria. The GFB is composed of three lay- Nephrin is specifically expressed by podocytes and is
ers, the specialized fenestrated endothelium, the glomerular involved in slit diaphragm formation and in regulating
basement membrane (GBM) and the podocyte. Damage to cellular functions; the loss of the proper function of this
any of these components can result in nephrotic proteinuria protein determines an early onset (usually during preg-
in various types of NS, but the podocyte is now recognized nancy or in the first months after birth) of massive pro-
as the key component in maintaining the anatomical and teinuria and NS, inevitably leading to ESRD. Following
functional integrity of the GFB. Hence, SRNS could be this discovery, the hunt for other possible genes responsible
defined as a structural podocytopathy, where genetic for NS received a great impulse. Mutations in NPHS2
alterations play a major role [2, 3, 11]. encoding for another slit diaphragm protein, podocin, were
In this review, we summarize the impact of the knowl- recognized as a relatively common etiology of autosomal
edge derived from the application of genetics to SRNS and recessive SRNS in childhood [20, 21]. Although mutations
we propose a new approach to children affected by this in NPHS1 and NPHS2 represent the most common genetic
disease. cause of SRNS in infancy and childhood, in the last two
decades the number of genes potentially causative for
SRNS has continuously increased [2, 22]. All of these
What did we know about SRNS? Historical notes genes encode for proteins that are expressed by podocytes
and traditional knowledge and that are probably essential to maintain the anatomical
or functional integrity, since their loss of function deter-
Increasing evidence suggests that the podocyte is the cul- mines protein leakage from the GFB and the sequence of
prit in many glomerular diseases including NS and that a events that finally lead to glomerular scarring and FSGS.
loss of more than 30 % of podocytes induces the occur- As a direct consequence, patients with NS related to a
rence of glomerulosclerosis [11]. Different types of pri- genetic alteration of the podocyte structure are insensitive
mary insult lead podocytes to react with an apparently to corticosteroid treatment, since alterations in podocyte
stereotypic pattern that includes foot processes effacement, integrity are intrinsic and not affected by the drug. These
detachment from the GBM, and/or death, finally impairing discoveries led to a fundamental improvement in our
the GFB to function and determining the deposition of understanding of the pathophysiology of SRNS and of
extra-cellular matrix with the formation of glomerular scars podocyte biology, although this was considered as poten-
[1214]. Whatever the starting event inducing podocyte tially involving only a rare subset of patients until the
injury and loss, the road toward glomerulosclerosis and advent of extended genetic sequencing techniques.

123
J Nephrol

What do we now know about SRNS? Recent severity, suggesting that different mechanisms of podocyte
advances and pathogenic classification of SRNS injury may be involved. These phenotypic differences are
probably related to the time of nephron development and
Next-generation sequencing (NGS), also known as high- maturation when these genes are expressed and start to
throughput sequencing, is a method of sequencing that carry out their specific function. Therefore, genes that are
allows to simultaneously obtain a large amount of genetic critical for early glomerular development would probably
data in a single run of sequencing. Independently of the lead to early onset of clinical manifestations, while genes
specific aim (e.g. whole-exome sequencing, target rese- encoding for proteins that regulate functions of mature
quencing) and the technology used, NGS is a time- and podocytes determine a delayed onset of the clinical phe-
cost-effective strategy to unravel the genetic sequence of notype and frequently present a more variable penetrance.
more than one gene at a time, usually in more than one From a functional point of view, gene products can now be
patient. For this reason, NGS is also referred to as mas- grouped in complexes that are often characterized by
sive-parallel sequencing. These techniques were intro- specific sub-cellular location. These include glomerular slit
duced in the 1990s and have progressively substituted the membrane components and signaling transducing proteins
traditional Sanger sequencing for genetic analysis. The (e.g. proteins expressed by genes NPHS1, NPHS2, CD2AP,
advent of NGS, together with the possibility to study large CRB2, PLCE1, TRPC6, DGKE, CUBN, ANLN, TTC21B),
cohorts of patients and to establish precise genotypephe- transcription factors (e.g. WT1, PAX2, LMX1B, SMAR-
notype correlations, has led to an impressive improvement CAL1, WDR73, NUP107), actin-binding proteins (ACTN4,
in our understanding of the pathogenesis of SRNS, allow- MYO1E, MYH9, INF2), integrins (e.g. ITGA3, ITGB4),
ing us to identify specific groups of patients that could GMB signaling components (e.g. LAMB2), actin-regulating
therefore benefit from a specific clinical approach. This small GTPases (e.g. ARHGAP24, ARHGDIA, KANK1, -2,
knowledge has prepared the ground for a critical revision -4), lysosomal proteins (e.g. SCARB2), proteins involved in
of the causative mechanisms of the disease. coenzyme Q10 biosynthesis (e.g. COQ2, COQ6, ADCK4,
PDSS2), and others (e.g. PTPRO) [2, 4].
The role of genetics Until the validation of NGS as a reliable diagnostic tool
for SRNS, the reported frequency of a genetic cause of the
About 10 years ago, by means of traditional Sanger disease was extremely variable, depending on the cohorts
sequencing technology, it was demonstrated that 85 % of and on the genes analyzed. This generated a tunnel view of
SRNS cases with onset by 3 months of age (congenital NS) the problem, inducing the inappropriate belief that genetic
and 66 % of cases with onset in the first year of life (in- forms of NS represented a rare subset that did not signifi-
fantile NS) could be explained by mutations in one of four cantly influence the therapeutic choices, which were usu-
genes (NPHS1, NPHS2, LAMB2 and WT1) [23]. Nowa- ally required well before the results of a genetic test could
days, the list of genes responsible for SRNS is considerably be available.
longer and continuously growing: besides classically rec- Indeed, traditional sequencing technology permits to
ognized SRNS-associated genes, such as NPHS1, NPHS2, study only single genes one at a time: it is therefore nec-
WT1 or LAMB2, a number of other genes have been essary to have a high suspicion about which specific gene
identified as causative of SRNS in children and even in might be responsible for the clinical phenotype of the
adults. They can be classified based on their pattern of patient before studying it. Nevertheless, a negative result
inheritance (autosomal recessive, autosomal dominant or does not exclude the genetic origin of the disease, since
X-linked), that frequently reflects in a different age of onset other genes that were not sequenced could anyway be
of the clinical phenotype and in different familial history. altered. Moreover, genetic testing was traditionally per-
While autosomal recessive forms of SRNS (the most formed in selected groups of patients with congenital or
common being caused by mutations in NPHS1, NPHS2, very early onset of clinical manifestations, or in familial
LAMB2 and PLCE1 genes) usually occur in infancy or clusters of the disease that represent genetically informa-
childhood and do not present familial grouping, genes tive populations and are characterized by a significant
transmitted with an autosomal dominant pattern of inheri- likelihood of a genetic-determined pathogenesis. These
tance (e.g. WT1, ACTN4, CD2AP, TRPC6) tend to present patients would benefit from a conservative therapy and
later in life, sometimes even in adulthood, and usually have from avoidance of immunosuppressive treatments. How-
familial aggregation, even though de novo mutations are ever, in every-day clinical practice the challenge is repre-
not infrequent, particularly for some genes (such as sented by sporadic cases of SRNS, since they are not
ACTN4). Moreover, they are characterized by a more distinguishable a priori from those who will respond to
varied penetrance with different degrees of disease steroid treatment solely on the basis of the clinical picture

123
J Nephrol

or even pathologic data coming from kidney biopsy [2, 24]. only 16 % of patients with genetic SRNS experienced a
Data on the frequency of a genetic cause in sporadic cases partial remission after CsA therapy. However, all genetic
of SRNS have rarely been reported and were frequently SRNS patients who had partial remission after treatment
mixed up with those of familial forms. Moreover, since no with CsA reached ESRD, suggesting that a reduction of
markers predicting a genetic defect were available, many proteinuria in those patients may represent a hemodynamic
patients used to undergo immunosuppressive treatment effect that does not mirror a true therapeutic effect [27].
until a clear diagnosis was established. Interestingly enough, the authors also reported that of those
However, evidence that genetic abnormalities account patients with renin-angiotensin aldosterone system inhibi-
for a significant proportion of patients affected by sporadic tors (RAAS-I) therapy without concomitant CsA (n = 24),
SRNS has recently appeared. It was demonstrated in a 54 % developed ESRD (n = 13), while in patients with
cohort of 69 children diagnosed with non-familial and non- RAAS-I and CsA therapy (n = 28), 71 % developed
syndromic NS [25] that, using NGS to target a panel of 46 ESRD (n = 20) [27]. Although the number of cases is
podocyte genes and a standardized protocol for variants limited and the difference is not significant, these results
prioritization, more than 30 % of children with a steroid- suggest that a prolonged treatment with CsA in patients
resistant phenotype showed heterogeneous genetic variants may even be harmful, considering the nephrotoxicity of the
that could be recognized as disease-causing. By contrast, drug. Taken together, these results, that other groups [22,
patients with SSNS did not present any pathogenic muta- 28] have also confirmed, show that a single-gene cause of
tion in the analyzed genes. The authors also demonstrated SRNS is anything but rare, even in sporadic cases. In fact,
that neither the age at onset of SRNS nor the pathologic if causative genes are considered and analyzed one at a
findings on renal biopsy are helpful in distinguishing time, they account for a negligible fraction of patients; in
patients with pathogenic variants from those who are contrast, by means of massive-parallel NGS strategies,
negative. More importantly, there was a significant corre- when genetic screening includes a larger group of causative
lation between the presence of genetic abnormalities and genes, a molecular diagnosis can be obtained in up to one-
the lack of response to immunosuppressive treatment, third of patients.
demonstrating that resistance to immunosuppressive treat-
ments in children with NS is associated with heterogeneous
genetic mutations in podocyte genes [25]. These data were Should we change our clinical approach
subsequently confirmed in a large cohort of 1783 patients to children with SRNS?
with SRNS manifesting before 25 years of age [26]. This
study identified a single-gene cause of SRNS in 29.5 % of The impressive burst in genetic knowledge about SRNS
patients, with a frequency of genetic diagnosis in sporadic raises the question of whether we can take advantage of the
cases of nearly 25 % which, even if less in comparison to information that comes from sequencing in order to modify
that found in consanguineous families (approximately our current protocols to ameliorate patient care [3]. Since
50 %), was nevertheless significant. Although the propor- patients with genetic forms of SRNS are known to be
tion of patients with a genetic disease decreased with unresponsive to immunosuppressive therapy, modifying
increasing age at onset, a significant fraction of a single- the intensity and duration of immunosuppression in indi-
gene cause was identified also in older children and in viduals with monogenic SRNS, sparing patients the unto-
young adults (more than 10 %), suggesting that a molec- ward side effects of prolonged ineffective treatments, is
ular diagnosis is not negligible even after infancy and early mandatory. We propose the following approach to children
childhood, as traditionally thought. Finally, a further study with idiopathic NS (Fig. 1): first, renal biopsy should be
from the German registry of pediatric patients reported an considered as soon as steroid-resistance becomes evident,
overall mutations detection rate as high as 41 % in patients in order to unravel possible rare different diagnoses (e.g.
with SRNS; 79 % of all mutations were identified by the membranous nephropathy or C3 glomerulopathy). If such
analysis of three single genes only (NPHS1, NPHS2, and cases are excluded, genetic testing should be immediately
WT1) [27]. Remission of the disease in non-genetic SRNS performed. In addition to patients with familial history,
was observed in 78 % of patients after a median treatment syndromic features and congenital onset of the disease, all
period of 2.5 months with calcineurin inhibitors, and 98 % patients with sporadic forms of NS failing to respond to an
of patients with non-genetic SRNS and cyclosporine A appropriate course of corticosteroid treatment (e.g. pred-
(CsA)-induced complete remission (normalbuminemia and nisone 60 mg/m2/day for 6 weeks, eventually followed by
no proteinuria) maintained a normal renal function over intravenous methylprednisolone, depending on the proto-
time. On the contrary, genetic SRNS was associated with a cols) should start a course of immunosuppressive treatment
high rate of ESRD (66 % of patients). Only one patient and simultaneously undergo extensive genetic testing with
with genetic SRNS experienced a complete remission and NGS strategies (target resequencing or whole-exome

123
J Nephrol

do not support the efficacy of immunosuppressive agents in


patients with proven genetic disease involving the podo-
cyte genes described to date [29], if the presence of
pathogenic mutations in one of these genes is observed in
the genetic testing, immunosuppressive therapy should be
interrupted and only anti-proteinuric treatment maintained
in order to preserve renal function and to avoid undesired
and potentially threatening side effects. On the other hand,
if genetic screening for mutations in such genes fails to
identify a cause of the disease, immunosuppressive treat-
ment should be continued, because the chance of a
response to immunosuppressive therapy is high [27]. This
may include anti-CD20 drugs (e.g. rituximab), even though
the long-term effects of this therapy are still far from being
completely assessed. In addition, while effective in patients
with forms of NS responsive to and even dependent on
steroids, rituximab and other anti-CD20 drugs have not
been compellingly demonstrated to have efficacy in
patients with SRNS, especially those who were multi-drug
resistant [30, 31]. Besides these consequences, genetic
testing can also provide other significant advantages.
Establishing a genetic diagnosis in SRNS can be of great
help in streamlining kidney transplant management, and
identifying the best donation option. In general, the precise
identification of a molecular diagnosis is of critical
importance for genetic counseling to the proband and to the
family. In addition, the assessment of a genetic cause of
Fig. 1 Flow chart illustrating the approach to children with nephrotic SRNS could help in reconsidering the presence of extra-
syndrome. In addition to patients with familial history, syndromic renal clinical findings of syndromic pictures that were not
features and congenital onset of the disease, all patients with sporadic immediately evident on clinical examination (e.g. pale
forms of NS failing to respond to an appropriate course of
bone abnormalities in LMX1B mutations) or in the evalu-
corticosteroid treatment should undergo an extended genetic screen-
ing with NGS. All the known SRNS causative genes should be ation of specific risks related to the molecular diagnosis
analyzed: NPHS1, NPHS2, CD2AP, CRB2, PLCE1, TRPC6, DGKE, (e.g. risk of developing Wilms tumor in patients with
CUBN, ANLN, TTC21B, WT1, PAX2, LMX1B, SMARCAL1, WDR73, genetic variants of WT1). Genetic screening may also guide
NUP107, ACTN4, MYO1E, MYH9, INF2, ITGA3, ITGB4, LAMB2,
in establishing effective specific therapies (e.g. coenzyme
ARHGAP24, ARHGDIA, KANK1, KANK2, KANK4, SCARB2, COQ2,
COQ6, ADCK4, PDSS2, PTPRO, FAT1, NUP93, NUP205, XPO5, Q10 supplementation in mutations of COQ10 genes or
SGPL1, AVIL, TUBAL3. NS nephrotic syndrome, CCS corticos- vitamin B12 supplementation in mutations of the CUBN
teroids, SRNS steroid-resistant nephrotic syndrome, SSNS steroid- gene) to delay or prevent renal function deterioration and
sensitive nephrotic syndrome, MCD minimal change disease, DMS
ameliorate extra-renal manifestations [3]. Finally, by col-
diffuse mesangial sclerosis, FSGS focal segmental glomerulosclero-
sis, CNIs calcineurin inhibitors, RAAIs renin-angiotensin-aldosterone lecting clinical and pathologic data, more precise geno-
system inhibitors typephenotype correlations can be established.

sequencing, depending on the availability). This implies


that patients should be referred to nephrology centers Conclusions and perspectives
where extended genetic screening can be performed with a
proven experience in data interpretation and application to Since the beginning of the 21st century, the availability of
clinical practice. While waiting for results of genetic test- NGS technologies has revolutionized the field of human
ing (that can be potentially available within a few weeks, genetics enabling high-throughput gene identification
depending on the technology used), a therapeutic course studies and introducing novel strategies to unravel the
with an immunosuppressive drug (usually calcineurin genetic etiology of Mendelian inherited disorders [32].
inhibitors) can be performed, together with anti-proteinuric This revolution has challenged the traditional view of
treatment (RAAS-I) and the response to such a therapy kidney disorders, including SRNS. Indeed, experimental
evaluated over a few months. However, since current data and clinical studies have largely demonstrated that

123
J Nephrol

Fig. 2 Etiology of steroid-


resistant nephrotic syndrome
(SRNS) across age groups.
Schematic representation of the
impact of different etiologic
factors of SRNS across age
groups, with a clear
predominance of genetic cause
in infancy, childhood and
adolescence. On the other hand,
environmental factors
(including infectious diseases,
drugs and immunologic
mechanisms) represent the main
cause of SRNS in increasing
age, although even in young
adults genetic causes are not
negligible

genetically-determined podocyte abnormalities are a pre- hyper- or hypo-activation of the RhoA pathway in podo-
viously underestimated cause of loss of permselectivity cytes may lead to a type of nephrotic syndrome that is
properties of the GFB (Fig. 2). NGS techniques (targeted instead partially sensitive to steroids [34], suggesting that
resequencing of selected genes, whole-exome and genome the type of genes involved can specifically predict the type
sequencing) have become progressively faster and cheaper, of response to treatment. These observations depict a (near)
allowing their application to clinical practice and to large- future where an extended genetic analysis of the patient
scale genetic studies. This has gradually led us to under- with NS will be essential to predict outcome and person-
stand the genetic background and cellular pathways alize treatments.
involved in SRNS, influencing the accuracy of prognosis, On the other hand, it must be considered that extensive
and guiding in treatment decisions and in genetic coun- genetic sequencing leads to the identification of an
seling. Moreover, as the response to treatment is the best impressive amount of variants that cannot be classified as
predictor of a patients prognosis in NS, finding parameters pathogenic but that may still play a role, even if sec-
that are predictive of response to therapy is essential in ondary, in the determination of the clinical phenotype of
order to avoid non-effective and potentially dangerous drug patients, that may even arise from primary immune injury
exposure. In SRNS patients, the genetic test can represent a or the presence of a circulating permeability factor. Thus,
suitable tool to distinguish those who will benefit from so-called modifier genes as well as di-genic and multi-
immunosuppressive treatments from those who will even- genic inheritance may deal with epigenetic events and
tually only risk the side effects related to such therapies. environmental effects [32]. This is the case of APOL1
However, the possibility to identify patients with the variants that represent risk alleles for FSGS in African
genetic form of SRNS is strictly dependent on a correct Americans: their identification by means of genome-wide
selection of patients to steer towards genetic testing. In association studies has opened new lines of investigation,
fact, while the selection of patients based on the clinical which may yield critical insights into the pathogenesis of
phenotype has been demonstrated to be highly informative the disease [4].
[25, 26], very different results have been obtained when In conclusion, SRNS is not a single disease entity but
other criteria of selection have been applied [33]: this is rather part of a spectrum of diseases with clearly defined
particularly true for biopsy findings, since the pathologic etiology, whose precise nature is progressively being elu-
pattern associated with the disease does not really distin- cidated [2]. Genetic information will expand our knowl-
guish between different etiologies [33]. edge about disease mechanisms involved in SRNS,
However, NGS and the advent of the genomic era of eventually leading to a better understanding of disease
kidney diseases have tracked routes that are still far from pathways and paving the way for the exploration of alter-
being definitively covered. As an example, very recent native pathogenic mechanisms, which may help in estab-
results suggest that variations in six different genes lishing a better clinical management of patients as well as
(MAGI2, TENC1, DLC1, CDK20, TLN1 and ITSN1) that in identifying new possible therapeutic targets for this, still
encode proteins of the Rho/Rac/Cdc42 network and induce as yet orphan, disease.

123
J Nephrol

Acknowledgments This article is supported by funding from the 17. Romagnani P, Remuzzi G (2013) Renal progenitors in non-dia-
Meyer Childrens Hospital Foundation and from the Fondazione betic and diabetic nephropathies. Trends Endocrinol Metab
AMARTI to Paola Romagnani. 24:1320
18. Lasagni L, Romagnani P (2010) Glomerular epithelial stem cells:
Compliance with ethical standards the good, the bad, and the ugly. J Am Soc Nephrol 21:16121619
19. Kestila M, Lenkkeri U, Mannikko M, Lamerdin J, McCready P,
Conflict of interest The results presented in this review have not Putaala H, Ruotsalainen V, Morita T, Nissinen M, Herva R,
been published previously in whole or part, except in the abstract Kashtan CE, Peltonen L, Holmberg C, Olsen A, Tryggvason K
format. All authors declare no conflict of interest. (1998) Positionally cloned gene for a novel glomerular protein
nephrinis mutated in congenital nephrotic syndrome. Mol Cell
Ethical approval This article does not contain any studies with 1:575582
human participants performed by any of the authors. 20. 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
References 24:349354
21. Brown EJ, Pollak MR, Barua M (2014) Genetic testing for
1. Eddy AA, Symons JM (2003) Nephrotic syndrome in childhood. nephrotic syndrome and FSGS in the era of next-generation
Lancet 362:629639 sequencing. Kidney Int 85:10301038
2. Lovric S, Ashraf S, Tan W, Hildebrandt F (2015) Genetic testing 22. Trautmann A, Bodria M, Ozaltin F, Gheisari A, Melk A, Azocar
in steroid-resistant nephrotic syndrome: when and how? Nephrol M, Anarat A, Caliskan S, Emma F, Gellermann J, Oh J, Baskin E,
Dial Transplant. doi:10.1093/ndt/gfv355 Ksiazek J, Remuzzi G, Erdogan O, Akman S, Dusek J, Davitaia
3. Saleem MA (2013) New developments in steroid-resistant T, O zkaya O, Papachristou F, Firszt-Adamczyk A, Urasinski T,
nephrotic syndrome. Pediatr Nephrol 28:699709 Testa S, Krmar RT, Hyla-Klekot L, Pasini A, O zcakar ZB, Sallay
4. Hall G, Gbadegesin RA (2015) Translating genetic findings in P, Cakar N, Galanti M, Terzic J, Aoun B, Caldas Afonso A,
hereditary nephrotic syndrome: the missing loops. Am J Physiol Szymanik-Grzelak H, Lipska BS, Schnaidt S, Schaefer F, Con-
Renal Physiol 309:F24F28 sortium, PodoNet (2015) Spectrum of steroid-resistant and con-
5. Gbadegesin R, Lavin P, Foreman J, Winn M (2011) Pathogenesis genital nephrotic syndrome in children: the PodoNet registry
and therapy of focal segmental glomerulosclerosis: an update. cohort. Clin J Am Soc Nephrol 10:592600
Pediatr Nephrol 26:10011015 23. Hinkes BG, Mucha B, Vlangos CN, Gbadegesin R, Liu J, Has-
6. Ehrich JMM, Brodehl J (1993) Long versus standard prednisone selbacher K, Hangan D, Ozaltin F, Zenker M, Hildebrandt F,
therapy for initial treatment of idiopathic nephrotic syndrome in Group, Arbeitsgemeinschaft fur Paediatrische Nephrologie Study
children. Arbeitsgemeinschaft fur Padiatrische Nephrologie. Eur (2007) Nephrotic syndrome in the first year of life: two thirds of
J Pediatr 152:357361 cases are caused by mutations in 4 genes (NPHS1, NPHS2, WT1,
7. [No author listed] (1981) International Study of Kidney Disease and LAMB2). Pediatrics 119:e907e919
in Children: The primary nephrotic syndrome in children. Iden- 24. Gipson DS, Troost JP, Lafayette RA, Hladunewich MA,
tification of patients with minimal change nephrotic syndrome Trachtman H, Gadegbeku CA, Sedor JR, Holzman LB, Moxey-
from initial response to prednisone. A report of the International Mims MM, Perumal K, Kaskel FJ, Nelson PJ, Tuttle KR, Bag-
Study of Kidney Disease in Children. J Pediatr 98:561564 nasco SM, Hogan MC, Dell KM, Appel GB, Lieske JC, Ilori TO,
8. Kidney Disease: Improving Global Outcomes (KDIGO) Sethna CB, Fervenza FC, Hogan SL (2016) Complete remission
Glomerulonephritis Work Group (2012) KDIGO clinical practice in the nephrotic syndrome study network. Clin J Am Soc Nephrol
guideline for glomerulonephritis. Kidney Int Suppl. 2:139274 11:8189
9. Hildebrandt F (2010) Genetic kidney diseases. Lancet 25. Giglio S, Provenzano A, Mazzinghi B, Becherucci F, Giunti L,
375:12871295 Sansavini G, Ravaglia F, Roperto RM, Farsetti S, Benetti E,
10. Haraldsson B, Nystrom J, Deen WMC (2008) Properties of the Rotondi M, Murer L, Lazzeri E, Lasagni L, Materassi M,
glomerular barrier and mechanisms of proteinuria. Physiol Rev Romagnani P (2015) Heterogeneous genetic alterations in spo-
88:451487 radic nephrotic syndrome associate with resistance to immuno-
11. Wiggins RC (2007) The spectrum of podocytopathies: a unifying suppression. J Am Soc Nephrol 26:230236
view of glomerular diseases. Kidney Int 71:12051214 26. Sadowski CE, Lovric S, Ashraf S, Pabst WL, Gee HY, Kohl S,
12. Lasagni L, Lazzeri E, Shankland SJ, Anders HJ, Romagnani P Engelmann S, Vega-Warner V, Fang H, Halbritter J, Somers MJ,
(2013) Podocyte mitosisa catastrophe. Curr Mol Med 13:1323 Tan W, Shril S, Fessi I, Lifton RP, Bockenhauer D, El-Desoky S,
13. Becherucci F, Lazzeri E, Romagnani P (2015) A road to chronic Kari JA, Zenker M, Kemper MJ, Mueller D, Fathy HM, Soliman
kidney disease: toward glomerulosclerosis via dendrin. Am J NA, SRNS Study Group, Hildebrandt F (2015) A single-gene
Pathol 185:20722075 cause in 29.5% of cases of steroid-resistant nephrotic syndrome.
14. Kriz W, Shirato I, Nagata M, LeHir M, Lemley KV (2013) The J Am Soc Nephrol 26:12791289
podocytes response to stress: the enigma of foot process 27. Buscher AK, Beck BB, Melk A, Hoefele J, Kranz B, Bam-
effacement. Am J Physiol Renal Physiol 304:F333F347 borschke D, Baig S, Lange-Sperandio B, Jungraithmayr T, Weber
15. Lasagni L, Angelotti ML, Ronconi E, Lombardi D, Nardi S, LT, Kemper MJ, Tonshoff B, Hoyer PF, Konrad M, Weber S,
Peired A, Becherucci F, Mazzinghi B, Sisti A, Romoli S, Burger (GPN), German Pediatric Nephrology Association (2016) Rapid
A, Schaefer B, Buccoliero A, Lazzeri E, Romagnani P (2015) response to cyclosporin A and favorable renal outcome in non-
Podocyte regeneration driven by renal progenitors determines genetic versus genetic steroid-resistant nephrotic syndrome. J Am
glomerular disease remission and can be pharmacologically Soc Nephrol 11(2):245253
enhanced. Stem Cell Reports 5:4863 28. Lipska BS, Iatropoulos P, Maranta R, Caridi G, Ozaltin F, Anarat
16. Remuzzi G, Benigni A, Remuzzi A (2006) Mechanisms of pro- A, Balat A, Gellermann J, Trautmann A, Erdogan O, Saeed B,
gression and regression of renal lesions of chronic nephropathies Emre S, Bogdanovic R, Azocar M, Balasz-Chmielewska I,
and diabetes. J Clin Invest 116:288296 Benetti E, Caliskan S, Mir S, Melk A, Ertan P, Baskin E, Jardim

123
J Nephrol

H, Davitaia T, Wasilewska A, Drozdz D, Szczepanska M, Jan- 31. Ravani P, Bonanni A, Rossi R, Caridi G, Ghiggeri GM (2016)
kauskiene A, Higuita LM, Ardissino G, Ozkaya O, Kuzma- Anti-CD20 antibodies for Idiopathic nephrotic syndrome in
Mroczkowska E, Soylemezoglu O, Ranchin B, Medynska A, children. Clin J Am Soc Nephrol 11(4):710720
Tkaczyk M, Peco-Antic A, Akil I, Jarmolinski T, Firszt-Adam- 32. Hardies K, Weckhuysen S, De Jonghe P, Suls A (2015) Lessons
czyk A, Dusek J, Simonetti GD, Gok F, Gheissari A, Emma F, learned from gene identification studies in Mendelian epilepsy
Krmar RT, Fischbach M, Printza N, Simkova E, Mele C, Ghig- disorders. Eur J Hum Genet. doi:10.1038/ejhg.2015.251 (Epub
geri GM, Schaefer F, Consortium, PodoNet (2013) Genetic ahead of print)
screening in adolescents with steroid-resistant nephrotic syn- 33. Sampson MG, Gillies CE, Robertson CC, Crawford B, Vega-War-
drome. Kidney Int 84:206213 ner V, Otto EA, Kretzler M, Kang HM (2015) Using population
29. Vivante A, Hildebrandt F (2016) Exploring the genetic basis of genetics to interrogate the monogenic nephrotic syndrome diagnosis
early-onset chronic kidney disease. Nat Rev Nephrol in a case cohort. J Am Soc Nephrol. doi:10.1681/ASN.2015050504
12(3):133146 34. Ashraf S, Rao J, Lawson JA, Tan W, Widmeier E, Airik M, Lovric
30. Magnasco A, Ravani P, Edefonti A, Murer L, Ghio L, Belingheri S, Wareijko J, Sadowski CE, Pabst W, Braun D, Jairajpuri MA, Gee
M, Benetti E, Murtas C, Messina G, Massella L, Porcellini MG, HJ, Lifton RP, Zenker M, Hildebrandt F (2016) Recessive mutations
Montagna M, Regazzi M, Scolari F, Ghiggeri GM (2012) in 5 novel genes of interaction partners elucidate steroid sensitivity
Rituximab in children with resistant idiopathic nephrotic syn- in nephrotic syndrome. In: 11th international podocyte conference,
drome. J Am Soc Nephrol 23:11171124 Haifa-Jerusalem, 36 April 2016

123

Das könnte Ihnen auch gefallen