Mechanisms of Glucocorticoid Resistance in Idiopathic Nephrotic Syndrome
Ruochen Che,Aihua Zhang
DOI: https://doi.org/10.1159/000350163
2013-01-01
Abstract:Nephrotic syndrome, characterized by massive proteinuria, hypoalbuminemia and edema, is one of the most common kidney diseases in children. Although glucocorticoids (GCs), the mainstay of therapy for over 50 years, are effective in most children, more than 20% develop GC resistant nephrotic syndrome (SRNS), among whom focal segmental glomerular sclerosis (FSGS) is a frequent pathological outcome and the cause of end-stage renal disease with a prevalence of 4% in the USA. Despite its clinical importance, the molecular basis of SRNS is unknown. In recent years, researchers have not only gained a new understanding of the roles of structural and functional abnormalities in GC receptors (GRs) in GC resistance, but have also gradually discovered close relationships between GC resistance in idiopathic nephrotic syndrome and podocyte-related molecules, like slit diaphragm (SD) molecules and so on. Here we mainly discussed these molecules and their physiological as well as pathological effects, including nephrin, podocin, CD2-associated protein (CD2AP), α-actinin-4, transient receptor potential cation channel 6 (TRPC6), phospholipase C epsilon-1 (PLCε1), Wilms' tumor suppressor gene 1 (WT1), Lmx1b, LAMB2, myosin 1e (MOY1E) and inverted formin 2 (IFN2). Mitochondrial cytopathies are also involved in GC resistance and well-reviewed [1,2], which will not be discussed in detail in this review. To those SRNS without any genetic defects, immunological disturbances are always involved and should be stressed. In this article, recent progress in research on the mechanisms of GC resistance in idiopathic nephrotic syndrome is reviewed.GRs are expressed in glomerular cells, such as podocytes [3], and translocate GCs to the nucleus [4]. The gene encoding the GR resides on chromosome 5q31-32 and includes 10 exons [5], among which exons 2-9 encode protein; exon 1, the 5'-untranslated region, plays an important role in cell type specific GR gene expression [5,6]. It is well known that different splicing, modification and transcription of genes generate various polypeptides. Following promoters A, B and C, exon 1 of the GR gene has three isoforms (A, B and C), and different pre-RNA splicing of exon 1A generates three different transcripts (1A1, 1A2 and 1A3). All five of these exon 1 isoforms connect to the same area of exon 2. Similarly alternatively spliced pieces of other exons produce different isoforms, such as GRα, GRβ, GRγ, GRA and GRP, of which the latter two are believed to be associated with the GC resistance phenotype. In addition, eight translation initiation sites on the GRα mRNA result in eight GRα isoforms (GRαA, B, C1-3 and D1-3). These GR isoforms have diverse cellular signal transduction potential [7,8].GRα is the only known receptor that binds GCs and has a positive transactivation potential [9]. When ligand-free, GRα forms a multi-protein complex with, for example, hsp90, hsp70, immunophilins, FKBPs, Cyp-40 and p23 in the cytoplasm [10,11]. The configuration of these complexes helps the receptor maintain a high affinity for GCs. On GC binding, GRα detaches from its chaperones and translocates to the nucleus, where GR homodimers bind to a cis-acting sequence including GC response elements (GREs) and negative GREs in the promoter region of target genes, leading to the activation or repression of transcription, respectively [8]. Furthermore, GRα modulates the signal pathway through mutual interaction with coactivator and corepressor transcription factors [7,8]. Coactivator molecules, such as CREB-binding protein and GR coactivator-2, activate the transcription of anti-inflammatory proteins. Corepressors, such as activator protein-1 (AP-1) and nuclear factor kappa B (NF-κB), undergo cross-talk with GR homodimers, which can attenuate the proinflammatory response mediated by these proteins.GRβ has the same first 727 amino acids as GRα, while GRα has an additional 50 amino acids [12]; thus, GRβ lacks the special GC-binding pocket possessed by GRα and fails to bind GCs [13,14]. It is known that the predominant role of GRβ is to exert a negative effect on GRα-induced transcriptional activity. Research [7,15,16] has indicated that overexpression of GRβ leads to imbalance of the GRα/GRβ ratio and may be an important cause of GC resistance in some patients with nephrotic syndrome. The mechanism of this can be summarized as follows. 1) GRβ competes for GRE-binding with GRα [17]. 2) GRβ forms a heterodimer with GRα. Its unique carboxyl-terminal 15 amino acids form two critical residues, L733 and N734 [18], that stabilize the dimer, which represses the transcriptional activity of GRα. 3) Recent reports [19] have shown that GRβ has its own transcriptional activity independent of GRα. Through its AF-1 domain, GRβ can lodge into the transcriptional complex formed by GRα and other cofactors and then repress transcription [19,20]. 4) It is hypothesized that GRβ directly modulates target gene transcription through “GRβ-specific response elements” [19].The signal transduction mediated by GRα can also be interrupted by GR modification by phosphorylation, nitrosylation or ubiquitination [7,8]. Phosphorylation of GRs can be achieved by several kinases, including extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) [21] and P38 mitogen-activated protein kinase (MAPK) [22], which are activated by microbial superantigens, inflammatory cytokines or macrophage migration inhibitory factor. Inflammatory cytokines also generate increased nitric oxide, leading to the nitrosylation of GRs [23]. JNK can directly activate the transcription factor AP-1, preventing GRs from binding to GRE or NF-κB [21], as discussed in detail below.GCs are conventionally considered to have immunosuppressive and anti-inflammatory effects. Actually, recent evidence [24] has shown that GCs may have a direct effect on podocytes in glomerular diseases through upregulating the expression of GRs. GCs are translocated to the nucleus by GRs, then modify gene expression to promote cell maturation and survival, stabilize the actin cytoskeleton and exert a positive effect on the key proteins nephrin, CD2AP, TRPC6, VEGF and IL-6. Not only do GCs prevent podocyte apoptosis [25], they also promote cell recovery from injury [26]. Additionally, Zhang et al discovered that GCs increase regeneration by augmenting the number of podocyte progenitors [27]. To FSGS, which is frequently resistant to GCs, studies have shown that circulating permeability factor is the main culprit that induces podocyte foot effacement and proteinuria. Serum soluble urokinase receptor (suPAR) is one of the critical permeability factor, contributing to actin cytoskeleton reorganization and SRNS [28,29]. Then calcineruin inhibitors, like cyclosporine A, emerge to be against proteinuria of FSGS patients and help to taper down the dose of GCs. It exerts the anti-proteinuric effect not only by immune-suppression, but also by inhibiting of uPAR signaling pathway and blocking dephosphorylation of synaptopodin, then, consequently, protecting podocyte integrity [29,30]. Therefore, it would be interesting to further study the mechanism of GCs sensitivity and resistance in nephrotic syndrome, aiming at developing more effective therapies.In podocytes and podocytic processes, podocyte-related molecules, like SD molecules, are important components of the glomerular filtration barrier. Numerous SD molecules expressed by podocytes have recently been discovered and have been shown to be associated with GC resistance in idiopathic nephrotic syndrome, including NPHSl, NPHS2, ACTN4, CD2AP, TRPC6, PLCεl, MOY1E and IFN2. Molecules related with transcriptional activity in podocytes, such as WT1 and LMXlB are also critical to podocyte integrity. Additionally, LAMB1, secreted by podocytes, contributes to the structure of GBM, should also be stressed.In SRNS, the genes encoding these molecules undergo various mutations, including homozygous and heterozygous missense, nonsense, frameshift, insertion, deletion and splice-site mutations, which result in structural and functional anomalies in the encoded proteins, and thereby to podocyte injury. Researchers have detected immature renal glomeruli in some children with SRNS, which might be a consequence of the mutation of cytoskeleton specific proteins. Thus, when mutation related with podocytes occurs, GCs may be unable to exert direct effects on disrupted podocytes, leading to GC resistance.NPHS1 is located on human chromosome 19q13.1 and includes 29 exons. It encodes the podocyte-expressed protein nephrin, a member of the immunoglobulin superfamily and a single-pass transmembrane protein comprising eight extracellular C2-type Ig-like domains, a fibronectin type III-like motif, a single transmembrane domain and a cytosolic C-terminal tail [31]. In vitro experiments suggest that nephrin is highly flexible and changes its conformation easily [32]. Nephrin constitutes the structural basis of the SD with other proteins [33,34]. It also plays an important role in signaling between podocytes [35].Mutations of NPHS1 lead to congenital nephrotic syndrome (CNS) of the Finnish type in an autosomal recessive manner. Children with CNS have massive proteinuria and severe edema, are usually GC resistant and die within 2 years after birth [36]. Recent studies have confirmed that mutations of NPHS1 also occur in people of non-Finnish origin and in all three types of SRNS (CNS and childhood- and adult-onset SRNS) [37,38]. The most prevalent mutations of CNS in the Finnish population include Finmajor (nt-121delCT, L41fsX91) and Finminor(c.3325C>T, R1109X), which were the first such mutations to be discovered [31]. To date, over 176 different mutations have been described [39]. Although the classical “Finnish type” mutation leads to GC resistance and progresses rapidly to end stage kidney disease, in several cases other NPHS1 mutations have been reported to be sensitive to GCs and the condition has not been so severe. In a worldwide cohort study of 42 non-Finnish cases of CNS [40], two patients exhibited a milder phenotype; one of them had a compound heterozygous truncated mutation in exon 10 and a missense mutation in exon 24. He achieved partial remission with GC treatment and maintained stable renal function. Kitamura [41] reported two cases of compound heterozygous missense mutations (C256R and V822M) in which remission of proteinuria was achieved without immunosuppressive treatment. Recently, two siblings with a homozygous missense mutation (c.1760T>G, p. L587R) showed partial remission with GC treatment [36]. Researchers [40] have speculated that the severity of the disease phenotype may be due to different gene mutations. Finmajor and Finminor lead to the absence of nephrin from the SD, causing severe SRNS. About 75% of missense mutations lead to misfolding of nephrin, which becomes trapped in the endoplasmic reticulum and fails to express at the plasma membrane [42]. A point mutation may disturb the location of ligand binding to the extracellular nephrin domains. R1160X, a nonsense mutation of exon 27, results in a milder CNS phenotype [43]. However, it remains difficult to clarify the correlations between genotype and phenotype, which is also influenced by, for example, gender and ethnicity. More work is required.NPHS2 maps to chromosome lq25-q31 and includes eight exons encoding an integral membrane protein named podocin. Podocin is a newly identified member of the stomatin family of lipid raft-associated proteins and comprises 383 amino acids with a single ‘hairpin-like' transmembrane domain. Its cytosolic C- and N-terminal domains are located in the cytoplasm. Podocin forms a homo-oligomeric complex that localizes to lipid rafts in the plasma membrane in the foot process of podocytes [44,45]. These lipid rafts contain many signal transduction molecules and recruit nephrin and CD2AP with podocin [46,47]. When the gene encoding podocin mutates, the distribution of nephrin and other key proteins in podocytes changes as a consequence. NPHS2 mutations were initially reported in 2000, in children with autosomal recessive familial CNS. About 10-28% of all non-familial childhood cases of SRNS are caused by podocin mutations. Almost all patients with two recessive mutations of NPHS2 are resistant to GC treatment. As mentioned above, different genotypes may influence the severity of disease. Kitamura et al. [48] reported a patient who was a compound heterozygote for the NPHS2 mutations R168C and P271L. An expression study showed that the R168C mutant tended to become trapped within the ER, whereas the P271L mutant reached the plasma membrane and retained partial function in anchoring the SD; as a consequence, the disease had a milder clinical course. In 2006, Franceschini et al. [49] identified four relatively common non-synonymous gene variants: R229Q, G34E, A61V and A242V. R229Q was once believed to be harmless because it occurs in 2-4% of healthy individuals [49]. However, further studies have shown that R229Q decreases podocin binding to nephrin [50]. Although it may have only a weak biological effect, it is now known that compound heterozygosity for R229Q and p.A284V maybe characteristic of late childhood- or adult-onset SRNS [50,51,52]. Santin et al. [53] also found that the phenotypes of late childhood- and adult-onset SRNS are more similar to each other than to early childhood-onset SRNS. Researchers have speculated that specific podocin mutations might determine the age of onset of SRNS and that R229Q might be an ancient mutation that has expanded by population migration. Experts [54,55,56] recommend that adults with FSGS, especially in European countries, should be screened for R229Q first; those who are carriers should be tested for a second mutation. Unnecessary GC treatment should never be administered to compound heterozygous patients.Located on chromosome 19q13, ACTN4 encodes α-actinin-4, the only member of the actinin family expressed in human podocytes [57]. Alpha-actinin-4 is an actin filament crosslinked protein with an actin-binding domain in the N terminus, four spectrin-like repeats in its center and two EF-hand motifs in the C terminus [58]. Alpha-actinin-4 is localized in contractile microfilaments within podocyte foot processes and contributes to the unique morphological features of the podocyte foot process by regulating molecule adhesion and cytoskeletal dynamics [59,60]. Mutations in ACTN4 are related to familial FSGS in an autosomal dominant manner [57,61]. There are two main etiological mechanisms associated with the disorders caused by the mutants: “gain of function” and “loss of function” [62]. Gain-of-function mutations promote actin binding and abnormal aggregation of α-actinin-4 leading to podocyte damage. Such mutations often occur in the actin-binding domain (e.g., K225E, K256E). K256E is the mouse mutant corresponding to human K255E [60]. The K256E mutation increases the affinity of α-actinin-4 for F-actin, which reduces the ability of α-actinin-4 to interact with other binding proteins, disrupting the normal cytoskeleton [63]. Furthermore, the mutant may induce proteotoxicity in podocytes, impair the proteins' function by misfolding them and eventually cause apoptosis [60]. Loss-of-function mutation was not fully recognized as a pathogenic factor in human nephropathy until 2011 [62]. Before this, animal experiments [64,65] had shown that ACTN4-absent mice exhibited abundant proteinuria with sclerosis of glomerular capillaries and podocyte foot process effacement, but whether humans were affected in the same way was unknown at that time. In 2011, Liu et al. [62] demonstrated that mutations in the non-actin-binding region (e.g., R310Q, Q348R) result in deficiencies of α-actinin-4 and its binding protein CLP36. This disruption of the α-actinin-4-CLP36 complex hinders RhoA signaling and the generation of traction force in podocytes. Clinically, ACTN4 mutations usually cause late childhood- and adult-onset nephrotic syndrome [57]. Choi et.al. [66] reported a heterozygous mutation (p.S262F) in 3- and 4-year-old siblings with FSGS. However, because the siblings also had an NPHS1 substitution, which mutation caused the disease could not be determined.The 80 kDa CD2AP encoded by chromosome 6p12 belongs to the immunoglobulin superfamily and serves as an intracellular ligand of T cell and natural killer cell CD2 receptors. It is widely expressed in almost all tissues except the brain and maintains stable connections between T cells and antigen-presenting cells. CD2AP has three Src homology 3 (SH3) domains at the NH2 terminus followed by a proline-rich region containing SH3-binding sites; its coiled-coil domain is located at the COOH terminus. The SH3 domains anchor CD2 by identifying the proline-rich sequence in the intracellular C terminus of CD2. The COOH terminus mediates the protein-protein binding and participates in cytoskeleton remodeling [67]. Expressed in podocytes, CD2AP interacts with nephrin and podocin, maintaining the integrity of podocytes and SDs. These three proteins are interdependent on each other to maintain their structure and location [46]. CD2AP also participates in cell signaling pathways through stimulating the p85 regulatory subunit of phosphoinositide 3-OH kinase-dependent activation of the serine-threonine kinase AKT [68]. TGF-β induces these anti-apoptotic pathways via CD2AP adaptor protein. In the absence of CD2AP, the proapoptotic p38 MAPK pathway mediated by TGF-β is over-activated and accelerates podocyte apoptosis [69,70]. Fully knocked-out mice (CD2AP-/-) exhibited extensive foot process effacement and died of a disease resembling human nephrotic syndrome at approximately 6 weeks after birth [34]. Heterozygous CD2AP+/- mice had abnormal glomeruli similar to the pathological changes seen in human FSGS [71]. Researchers [72] have also reported that CD2AP can bind dendrin, a transcriptional factor. Loss of CD2AP leads to the release of dendrin and increased expression of TGF-β1, which drives translocation of dendrin from the SD to the nucleus. Dendrin promotes expression of cytosolic CatL, which can reorganize the actin cytoskeleton to make podocytes sensitive to proapoptotic signals. Thus, CD2AP, deletion of which may mediate a proteolytic process, plays an important role in cell survival. Kim et al. [73] reported a heterogeneous mutation of exon 7 in two patients with primary FSGS; the mutation replaced cytosine with thymidine, leading to a lack of splicing at that site and loss of 80% of CD2AP protein. A homozygous mutation of CD2AP leading to a premature stop codon has also been described, resulting in a slightly truncated protein. This mutation downregulated the expression of CD2AP and was discovered in a case of early-onset nephrotic syndrome [74]. Gigante et al. [75] screened for mutant CD2AP genes in 80 Italian sporadic FSGS patients and 200 healthy controls. Three mutations (c.904A>T, c.1120A>G and c.1573delAGA) were discovered in three unrelated patients who exhibited proteinuria and a high degree of GC resistance.TRPC channels are a superfamily comprising seven different channels divided into four subfamilies (TRPC1, TRPC3,6,7, TRPC4,5 and TRPC2) [76]. Localized on chromosome 11q21-22, TRPC6 encodes an important Ca2+ channel in podocytes [77]. This 100 kDa protein has both its N and C termini located intracellularly and contains six transmembrane domains, the fifth and sixth of which form tetramers [76]. Most TRPCs are nonselective cation channels, but TRPC6 is a selective ion channel [78]. Independent of intracellular calcium concentration and membrane depolarization, TRPC6 can be activated by PLCε1 after the stimulation of a G(q)-protein-coupled or tyrosine kinase receptor; this is followed by the release of intracellular calcium from the endoplasmic reticulum [76]. The entrance of Ca2+ promotes the assembly and reorganization of actin, responsible for cell migration and endothelial permeability [79,80]. TRPC6 is expressed throughout the kidney and specifically in the major and minor processes of podocytes. It also interacts with nephrin, podocin and CD2AP [81,82]. Overexpression of TRPC6 results in certain glomerular diseases via podocyte dysfunction caused by abnormal Ca2+ reflux [83]. Recently, Yu et al. [78] showed that dexamethasone stabilizes the expression of TRPC6 by binding its receptor, which protects podocytes from injury and plays a role in preventing proteinuria. However, the mutant TRPC6 seems to reduce the effect of GCs. Winn et al. [77] showed that a missense mutation (P112Q) of TRPC6 is related to hereditary FSGS. Gigante M et al. [84] analyzed TRPC6 in 33 Italian children with sporadic early-onset SRNS. Three heterozygous missense mutations (c.374A>G, c.653A>T and c.2684G>T) were recognized. In addition, Santin et al[85] identified three missense substitutions in non-familial cases. Mir et al. [86] detected a L395A missense variant in a sporadic FSGS patient. However, the precise genotype-phenotype relationship remains unknown. One hypothesis is that calcium overload activates calcium-dependent phosphatase calcineurin [83,87]. FK506 and CsA, which are required by GC-resistant patients, exert their effect by inhibiting calcineurin [88,89]. Therefore, mutations in TRPC6 might be correlated with GC resistance and the inhibition of calcineurin.The PLCε1 gene is on chromosome 10q23 and encodes PLCε1, a member of the phospholipase enzyme family [90,91]. PLC enzymes are divided into four classes: PLCβ, PLCγ, PLCδ and PLCε [92]. PLC works as a catalyst that promotes the hydrolysis of polyphosphoinositides to generate the second messengers, inositol-1,4,5 triphosphate (IP3) and diacylglycerol (DAG). IP3 releases Ca2+ from the endoplasmic reticulum while DAG is responsible for protein kinase C stimulation, both of which initiate cell growth and differentiation [91]. PLCε1 is highly expressed in podocytes and contains not only the conserved PLC part, but also a domain of guanine nucleotide exchange factor for Ras-like small GTPases and two C-terminal Ras-binding domains, through which it can be regulated by H-Ras. It can also interact with human BARF (γ-raf murine sarcoma viral oncogene homolog B1) and GTPase-activating protein 1, which is located in the basal part of developing podocytes [90,91,93]. Scientists [94] have found that PLCε1 mutation is a novel cause of DMS, which is characterized by the onset of nephrotic syndrome and rapid progression to end stage renal failure. Some patients presenting with nephrotic syndrome during the first year of life exhibit GC resistance [95]. Recently, a large cohort study of patients with isolated DMS showed that 28.6% of families with DMS have loss-of-function mutations in PLCε1. Eight of the ten identified families are consanguineous [96]. Rasheed et al. [97] reported eight different mutants with a late onset of FSGS, two of which were novel variants (R561Q and K2173R).Myo1e locates in chromosome 15q21-q22, encoding a member of the nonmuscle class I myosins that belong to a subgroup of the unconventional myosin protein family. Myo1e is an actin-based molecular motor that contributes to junctional integrity in kidney podocytes. Myo1e is composed by an N-terminal motor domain, a neck domain and a tail domain. The N-terminal domain is responsible for actin binding and ATPase activity. The tail domain contains three tail homology (TH) regions that bind acidic phopholipids and prolin-rich proteins[98]. Myo1e-null mice exhibit glomerular filtration defects and, consequently, extensive proteinuria, of which the mechanism may involve changes of podocyte adhesion and cytoskeletal organization rather than loss of other podocyte-related molecules [99,100]. Bi et al demonstrated that myo1e localizes in cell-cell junctions and its TH3 domain interacts with a component of SD and tight junctions, ZO-1 [98], contributing to the structure and function of SD. Simone et al. utilized homozygosity mapping and exome sequencing to identify an A159P substitution in MYO1E in three siblings with steroid-unresponsive FSGS. A159P mutation leads to molecular mislocation to the cytoplasm, impairing ligand binding and actin interaction [101]. Almost the same time, Mele et al. reported two mutations (A159P and Y695X) in MYO1E resulted in autosomal recessive GC resistant FSGS. Y695X mutation truncated the protein, lacking the domain for regulation of actin binding and ATP hydrolysis, which mimics the animal model of Myo1e deficiency. Cyclosporine A was partially effective in spite of high relapse rate [102]. To date, MYO1E mutations were homozygous and only reported in familial FSGS with some degree of consanguinity. So MYO1E mutations should be screened out in patients with familial FSGS.Locating on chromosome 14q32.33, INF2 encodes a member of the forming family of proteins. Formins remodel actin cytoskeleton and govern dynamic events like cell morphogenesis and cytokinesis [103]. INF2 regulates lamellipodial actin dynamics and SD trafficking by inhibiting actin polymerization mediated by Rho/diaphanous-related formins and interacting with lipid raft components [104]. Diaphanous-related formins contain the forming homology domains FH1/FH2 and the diaphanous autoregulatory domain (DAD) in C terminal whereas the diaphanous inhibitory domain (DID) is located in the N-terminal. FH1 and FH2 are responsible for acceleration of filament elongation and actin assembly, respectively, of which the latter can be inhibited by DID and DAD interaction. Rho GTPases can relieve the inhibition. However, DID-DAD interaction does not influence INF2 polymerization. In contrast, INF2 depolymerization is inhibited by the competion for actin monomer binding of DID-DAD [105]. Mutations in INF2 have been reported to be a major cause of autosomal dominant FSGS while sporadic cases are rarely seen. Barua et al concluded INF2 mutations reported in the literature [106]. Additionally, IFN2 mutations cause many cases of Charcot-Marie-Tooth syndrome (CMT) associated with FSGS. INF2 strongly expressed in podocytes and Schwann cell cytoplasm. All mutations detected are located in the DID domain and mutations reported in the cases of FSGS with CMT are concentrated in the inner face of the central core of DID. The mutations severely disrupt DID function, affecting its depolymerization effect and the interaction of INF2 with myelin-specific proteins [107,108], which might result in abnormal actin accumulation and INF2 dislocation [105,109] . Further studies about the mechanisms about renal and neural defects induced by INF2 mutations are expected.Wilms' tumor was first described by Max Wilms in 1899. It is known as a highly malignant nephroblastoma [110]. Investigators first identified constitutional deletions of the 11p13 chromosome in children with Wilms' tumor in 1978 [111]. In 1990, Haber et al. [112] isolated the Wilms' tumor suppressor gene WT1 by deletion analysis. WT1 has been recognized as a tumor-suppressor gene, but the wild type is often overexpressed in various cancers. The WT1 gene has 10 exons encoding a protein with four zinc fingers and a proline- and glutamine-rich transactivation domain, the structure of which is similar to that of transcription factors. The first exon encodes the transactivation domain alone, while exons 7-10 encode the four zinc fingers responsible for DNA and RNA binding [113]. During nephrogenesis, the WT1 protein plays an important role in the induction of the mesenchymal-epithelial transition and the formation of nephrons [114]. After kidney maturation, WT1 is expressed within podocytes [115]. The WT1 gene is spliced at the 17AA and KTS sites, yielding four basic isoforms (17AA(+)KTS(+), 17AA(+)KTS(-), 17AA(-)KTS(+) and 17AA(-)KTS(-)), each of which has different functions [116]. The 17AA(-)KTS(-)WT1 isoform inhibits G1/S progression during the cell cycle, suggesting a tumor-suppressor role, while Ito et al. [116] have shown that the 17AA(+)WT1 isoforms (both KTS(+) and KTS(-)) disturb apoptosis through the intrinsic apoptosis pathway. Thus, imbalanced expression of different WT1 isoforms results in abnormal podocyte hyperplasia and abnormal differentiation, inducing renal inflammation or tumor [117]. WT1 can also modify the cytoskeleton of podocytes through downregulating target genes encoding key podocyte proteins [118,119].There are three main nephropathies associated with WT1 mutation. The first is Denys-Drash syndrome (DDS), characterized by SRNS with diffuse messangial sclerosis (DMS), XY pseudohermaphrodism and Wilms' tumor [115]. Over 80 mutations have been reported in patients with DDS, most of which are missense in exon 8 or 9 encoding zinc fingers 2 and 3 [120]. The most common mutation is R394W (1180C>T, exon 9). It has been shown that, in the podocytes of DDS patients, WT1 mutants result in overexpression of Pax-2, which should be repressed after early nephrogenesis [110]. Second, mutations in the donor splice site at intron 9 lead to Frasier syndrome (FS), which is described as a combination of complete XY gonadal dysgenesis, FSGS and gonadoblastoma in 46, XY patients and as nephropathy alone in 46, XX patients [121]. The third nephropathy is isolated SRNS. Carrying the same mutation as that in DDS patients, some affected individuals exhibit only the isolated clinical manifestations of SRNS. This phenotype is usually found in females or males with genitourinary malformations [120,122].Encoding by Lmx1b, which is located on chromosome 9q34, LMX1B belongs to the LIM-homeodomain family of more than nine transcriptional factors. It contains two cysteine-rich N-terminal zinc-binding LIM domains, one homeodomain and a C-terminal glutamine-rich domain. The LIM domains interact with other transcription factors or modifiers, whereas the homeodomain is responsible for the binding of promoters [123
What problem does this paper attempt to address?
-
The β isoform of GSK3 mediates podocyte autonomous injury in proteinuric glomerulopathy.
Changbin Li,Yan Ge,Lance Dworkin,Ai Peng,Rujun Gong
DOI: https://doi.org/10.1002/path.4692
2016-01-01
Abstract:Converging evidence points to glycogen synthase kinase (GSK) 3 as a key player in the pathogenesis of podocytopathy and proteinuria. However, it remains unclear if GSK3 is involved in podocyte autonomous injury in glomerular disease. In normal kidneys, the isoform of GSK3 was found to be the major GSK3 expressed in glomeruli and intensely stained in podocytes. GSK3 expression in podocytes was markedly elevated in experimental or human proteinuric glomerulopathy. Podocyte-specific somatic ablation of GSK3 in adult mice attenuated proteinuria and ameliorated podocyte injury and glomerular damage in experimental adriamycin (ADR) nephropathy. Mechanistically, actin cytoskeleton integrity in podocytes was largely preserved in GSK3 knockout mice following ADR insult, concomitant with a correction of podocyte hypermotility and lessened phosphorylation and activation of paxillin, a focal adhesion-associated adaptor protein. In addition, GSK3 knockout diminished ADR-induced NFB RelA/p65 phosphorylation selectively at serine 467; suppressed de novo expression by podocytes of NFB-dependent podocytopathic mediators, including B7-1, cathepsin L, and MCP-1; but barely affected the induction of NFB target pro-survival factors, such as Bcl-xL. Moreover, the ADR-elicited podocytopenia and podocyte death were significantly attenuated in GSK3 knockout mice, associated with protection against podocyte mitochondrial damage and reduced phosphorylation and activation of cyclophilin F, a structural component of mitochondria permeability transition pores. Overall, our findings suggest that the isoform of GSK3 mediates autonomous podocyte injury in glomerulopathy by integrating multiple podocytopathic signalling pathways. Copyright (c) 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
-
Significance of Α-and Β-Isoforms of Glucocorticoid Receptors Expression in Children with Idiopathic Nephrotic Syndrome
Xiaojie He
2001-01-01
Abstract:Objective Glucocorticoid (GC) is the principal therapeutic drug in the treatment of idiopathic nephrotic syndrome (INS), and the response to GC treatment is an important indicator for the outcome of INS children. Children with GC-resistant INS are usually incompletely or non responseive to GC, and may herald the progression to end-stage renal failure. However, the detailed mechanism for why some INS children respond to GC and others do not, have still not been clearly elucidated. It is well known that GC action is mediated by the GC receptor (GR), a ligand-dependent transcription factor that belongs to the super-family of nuclear receptors. Two human GR isoforms termed GR α and GR β have been described. Recent studies demonstrated that the β-isoform of GR (GR β) is a potential endogenous inhibitor of the GC action in humans. So far, no research papers have involved in the relationship between the levels of two GR isoforms, GR α and GR β, expression in INS children and the response to GC in INS children. The aim of the present study was to determine the levels of GR α and GR β in INS children with the variable response to the GC treatment. Methods Twenty patients aged 4~13 years with INS were classified as GC-sensitive INS (8 patients) or GC-resistant INS (12 patients), according to their response to an eight-week course of oral prednisone. The heparinized venous blood was collected, and peripheral blood mononuclear cells (PBMC) were isolated by Ficoll-Hypaque gradient centrifugation. The immunocytochemical assay was used to investigate the expression of two isoforms of GR in PBMC. The cytoplasm protein and nuclear protein extracts from PBMC were obtained, and the quantity of GR α and GR β proteins were analyzed by Western blotting assay. Then the GR-DNA-binding activity was detected by electrophoretic mobility shift assay (EMSA). Results The two isoforms of GR were expressed in PBMC in INS and healthy children. The percentages of GR αpositive staining cells in GC-resistance INS and GC-sensitive INS were significantly lower than that in healthy children (P0.01), but no significant difference was found between two INS groups. However, the number of GR β positive staining cells in GC-resistant INS was significantly higher than that in GC-sensitive INS (P0.01). Results of Western blots assay identified the presence of only one specific protein band at the expected molecular weight 94 000 or 90 000, which was consistent with those of GR α and GR β, respectively. In GC-resistant INS children, the quantity of GR α protein in GC-resistant INS group was significantly higher in the cytoplasm, and significantly lower in the nuclei, while the quantity of GR β protein in the nuclei was significantly higher than that of GC-sensitive INS, respectively. The results of EMSA showed that GR-DNA-binding activity in GR-resistant INS group was decreased compared to GR-sensitive INS group. Conclusions The patients with GC-resistant INS have an increased expression of GR β. The imbalanced expression between GR α and GR β, especially increased expression of GR β in PBMC might account for GC-resistant INS. GR β might antagonize GC-mediated GR α function by prohibiting the GR α translocation into the nuclei and depressing GR-DNA binding activity. This finding suggests that GR β might be a critical factor regulating the responsiveness of the target cell to GC. The ability of GR β to repress the action of GR α also illustrates a new insight to the satisfactory explanation of why some INS children respond to GC and others do not.
-
[Effect of Glucocorticoid on Glucocorticoid-Resistant Children with Primary Nephrotic Syndrome].
Xiao-jie He,Zhu-wen Yi,Xi-qiang Dang,Hui-qiong Zhang,Qing-nan He,Shuang-hong Mo,Hai-tao Bai,Wen-mao Geng,Hua-bin Yang
DOI: https://doi.org/10.3760/j.issn:0578-1310.2005.02.012
2005-01-01
Abstract:OBJECTIVEGlucocorticoid (GC) is the first therapeutic choice of primary nephrotic syndrome (PNS). The response to GC treatment is an important indicator for the outcome of PNS children. Children with GC-resistant PNS present with incomplete or no response to GC, and may herald the progression to end-stage renal failure. However, the detailed mechanism of GC-resistance or GC-sensitive effect in these PNS children has not been clearly elucidated. The previous study by the authors indicated that there was increased expression of GR beta in PBMCs in GC-resistant children with PNS, and the over expression of GR beta resulted in GC resistance via influencing the ability of GR alpha nuclear translocation. To elucidate the relationship between GR beta expression in renal and in PBMCs and the effect of glucocorticoid on glucocorticoid-resistance children with PNS, the expression of GR alpha and GR beta in renal tissue and in PBMCs were detected by immunohistochemistry.METHODSForty children with PNS were divided into two groups, GC-resistant group(20) and GC-sensitive group(20), the expression of GR alpha and GR beta in renal intrinsic cells and in PBMCs were measured with the immunohistochemistry technique. A semiquantitative score was used to evaluate the injury degree of the glomeruli and tubulointerstitium.RESULTSCompared with GC-sensitive group, the glomerular pathologic scores (6.91 +/- 1.98) and renal tubular pathologic scores (7.12 +/- 1.62) in GC- resistant group were significantly different (P < 0.01, respectively). GR alpha expressions of renal tissue and PBMCs were higher in the control group (58.3 +/- 2.6, 59.1 +/- 7.2) than those in the GC-sensitive group (40.2 +/- 7.2 and 36.6 +/- 5.1, P < 0.01, respectively) and GC-resistant group (35.0 +/- 8.2 and 36.4 +/- 6.6, P < 0.01, respectively). GR beta expressions of renal tissue and PBMCs were higher in the GC-resistant group (13.8 +/- 3.0 and 12.1 +/- 4.1) and in the GC-sensitive group (6.5 +/- 1.9 and 5.9 +/- 1.0) than that in control group (2.3 +/- 0.4 and 3.2 +/- 1.1, P < 0.01, respectively). GR beta expressions in renal tissue and PBMCs were higher in the GC-resistant group than that in the GC-sensitive group (P < 0.01). Compared with control group, GR beta expressions in PBMCs and in renal tissue were lower than those in mild renal lesion group (5.4 +/- 2.8, 6.46 +/- 2.50), midmedium renal lesion group (8.7 +/- 2.4 and 11.4 +/- 3.7) and (17.1 +/- 0.4 and 18.7 +/- 0.7) in severe renal lesion group (F = 5.8, 15.6, P < 0.01, respectively). GR beta expression of PBMCs had a positive correlation with GR beta expression of renal intrinsic cells (r = 0.651, P < 0.01). GR beta expressions by PBMCs and renal intrinsic cells were positively correlated with renal pathologic scores (r = 0.579 and 0.623, P < 0.01, respectively).CONCLUSIONGC-resistant children with PNS were related to the increased GR beta expression in PBMCs and renal intrinsic cells. There was no correlation between the GR alpha expressions in PBMCs and in renal intrinsic cells. Increased GR beta expression might decrease the effect of GC via inhibiting the activity of GR alpha.
-
Glomerular Transcriptome Profiles in Focal Glomerulosclerosis: New Genes and Pathways for Steroid Resistance.
Jun Tong,Yuanmeng Jin,Qinjie Weng,Shuwen Yu,Hafiz Muhammad Jafar Hussain,Hong Ren,Jing Xu,Wen Zhang,Xiao Li,Weiming Wang,Jingyuan Xie,Nan Chen
DOI: https://doi.org/10.1159/000505956
2020-01-01
American Journal of Nephrology
Abstract:Background:Patients with focal segmental glomerulosclerosis (FSGS) characterized by steroid-resistant nephrotic syndrome (SRNS) are prone to progress to ESRD. Mechanism for the FSGS patients' response to steroid treatment is still unknown and currently, it is impossible to predict the steroid resistance before treatment of patients with FSGS.Methods:To identify biomarkers and potential therapeutic targets of FSGS patients with SRNS, patients diagnosed as kidney biopsy-proven FSGS and nephrotic syndrome (NS) were prospectively enrolled. They were divided into 2 groups, steroid-sensitive NS and SRNS based on their treatment response. Cortical regions were selected from biopsied renal tissues, and glomeruli were isolated under an inverted microscope. RNA was prepared from the isolated glomeruli and further used for microarray analysis. Followed by multiple analyses, the top 6 highest and lowest, and a selected panel of differentially expressed genes obtained and their related pathways were validated via real-time PCR, western blot, and measurement of reactive oxygen species (ROS).Results:In SRNS group, we discovered that the most significant up-regulated pathway was primarily related to cellular amino acid and derivative metabolic process. Meanwhile, the most significant down-regulated pathway was primarily involved in anatomical structure morphogenesis. Moreover, we foundNADPHoxidase 4(NOX4), one of the key regulators of renal ROS, at a much higher level in SRNS both at transcriptomic and proteomic levels. We also found the levels of ROS, p-p38 MAPK and matrix metalloproteinase (MMP)-2, which were all regulated by NOX4, were also higher in glomeruli isolated from SRNS patients. At last, we detected stimulated by retinoic acid gene 6 homolog (STRA6), a cell surface receptor formerly known as a gene preventing podocytes from over-proliferative lesion induced by HIV infection and was up-regulated by retinoic acid, expressed at a much higher level in SRNS kidneys.Conclusion:We found 2 potential mechanisms underline the SRNS, NOX4/ROS/P38 MAPK/MMP-2 pathway and STRA6. Our findings provided new insights into the steroid resistance.
-
Genetic and Pharmacologic Targeting of Glycogen Synthase Kinase 3 Beta Reinforces the Nrf2 Antioxidant Defense Against Podocytopathy
Sijie Zhou,Pei Wang,Yingjin Qiao,Yan Ge,Yingzi Wang,Songxia Quan,Ricky Yao,Shougang Zhuang,Juan Wang,Yong Du,Zhangsuo Liu,Rujun Gong
DOI: https://doi.org/10.1681/asn.2015050565
IF: 14.978
2016-01-01
Journal of the American Society of Nephrology
Abstract:Evidence suggests that the glycogen synthase kinase 3 (GSK3)-dictated nuclear exclusion and degradation of Nrf2 is pivotal in switching off the self-protective antioxidant stress response after injury. Here, we examined the mechanisms underlying this regulation in glomerular disease. In primary podocytes, doxorubicin elicited cell death and actin cytoskeleton disorganization, concomitant with overactivation of GSK3 beta (the predominant GSK3 isoform expressed in glomerular podocytes) and minimal Nrf2 activation. SB216763, a highly selective small molecule inhibitor of GSK3, exerted a protective effect that depended on the potentiated Nrf2 antioxidant response, marked by increased Nrf2 expression and nuclear accumulation and augmented production of the Nrf2 target heme oxygenase-1. Ectopic expression of the kinase-dead mutant of GSK3 beta in cultured podocytes reinforced the doxorubicin-induced Nrf2 activation and prevented podocyte injury. Conversely, a constitutively active GSK3 beta mutant blunted the doxorubicin-induced Nrf2 response and exacerbated podocyte injury, which could be abolished by treatment with SB216763. In murine models of doxorubicin nephropathy or nephrotoxic serum nephritis, genetic targeting of GSK3 beta by doxycycline-inducible podocyte-specific knockout or pharmacologic targeting by SB216763 significantly attenuated albuminuria and ameliorated histologic signs of podocyte injury, including podocytopenia, loss of podocyte markers, podocyte de novo expression of desmin, and ultrastructural lesions of podocytopathy (such as foot process effacement). This beneficial outcome was likely attributable to an enhanced Nrf2 antioxidant response in glomerular podocytes because the selective Nrf2 antagonist trigonelline abolished the proteinuria-reducing and podocyte-protective effect. Collectively, our results suggest the GSK3 beta-regulated Nrf2 antioxidant response as a novel therapeutic target for protecting podocytes and treating proteinuric glomerulopathies.
-
Fine-tuning of NFκB by Glycogen Synthase Kinase 3β Directs the Fate of Glomerular Podocytes Upon Injury.
Hui Bao,Yan Ge,Ai Peng,Rujun Gong
DOI: https://doi.org/10.1038/ki.2014.428
IF: 19.6
2015-01-01
Kidney International
Abstract:Nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) is regulated by a myriad of signaling cascades including glycogen synthase kinase (GSK) 3β and plays a Janus role in podocyte injury. In vitro, lipopolysaccharide (LPS) or adriamycin (ADR) elicited podocyte injury and cytoskeletal disruption, associated with NFκB activation and induced expression of NFκB target molecules, including pro-survival Bcl-xL and podocytopathic mediators like MCP-1, cathepsin L, and B7-1. Broad-range inhibition of NFκB diminished the expression of all NFκB target genes, restored cytoskeleton integrity, but potentiated apoptosis. In contrast, blockade of GSK3β by lithium or 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8) mitigated the expression of podocytopathic mediators, ameliorated podocyte injury, but barely affected Bcl-xL expression or sensitized apoptosis. Mechanistically, GSK3β was sufficient and essential for RelA/p65 phosphorylation, specifically at serine 467, which specifies the expression of selective NFκB target molecules, including podocytopathic mediators, but not Bcl-xL. In vivo, lithium or TDZD-8 therapy improved podocyte injury and proteinuria in mice treated with LPS or ADR, concomitant with the suppression of podocytopathic mediators, but retained Bcl-xL in glomerulus. Broad-range inhibition of NFκB conferred similar but much weakened antiproteinuric and podoprotective effects accompanied with a blunted glomerular expression of Bcl-xL and marked podocyte apoptosis. Thus, the GSK3β-dictated fine-tuning of NFκB may serve as a novel therapeutic target for podocytopathy.
-
Novel Variants in CRB2 Targeting the Malfunction of Slit Diaphragm Related to Focal Segmental Glomerulosclerosis
Qing Yang,Dan Tang,Chun Gan,Mi Bai,Xiaomei Song,Wei Jiang,Qiu Li,Yaxi Chen,Aihua Zhang,Mo Wang
DOI: https://doi.org/10.1007/s00467-023-06087-6
2024-01-01
Pediatric Nephrology
Abstract:Focal segmental glomerulosclerosis (FSGS) is a leading cause of steroid-resistant nephrotic syndrome (SRNS) that predominantly affects the podocytes. While mutations in genes causing pediatric SRNS have enhanced our understanding of FSGS, the disease’s etiology remains complex and poorly understood. Whole exome sequencing (WES) was performed on a 9-year-old girl with SRNS associated with FSGS (SRNS–FSGS). We analyzed the expression of CRB2, slit diaphragm (SD)-associated proteins, and sphingosine 1-phosphate receptor 1 (S1PR1) in the proband and CRB2 knock-down podocytes. In this study, we identified two novel compound heterozygous mutations in the Crumbs homolog 2 (CRB2) gene (c.2905delinsGCCACCTCGCGCTGGCTG, p.T969Afs*179 and c.3268C > G, p.R1090G) in a family with early-onset SRNS–FSGS. Our findings demonstrate that these CRB2 abnormalities were the underlying cause of SRNS–FSGS. CRB2 defects led to the dysfunction of podocyte SD-related proteins, including podocin, nephrin, and zonula occludens-1 (ZO-1), by reducing the phosphorylation level of S1PR1. Interestingly, the podocytic cytoskeleton remained unaffected, as demonstrated by normal expression and localization of synaptopodin. Our study also revealed a secondary decrease in CRB2 expression in idiopathic FSGS patients, indicating that CRB2 mutations may cause FSGS through a previously unknown mechanism involving SD-related proteins. Overall, our findings shed new light on the pathogenesis of SRNS–FSGS and revealed that the novel pathogenic mutations in CRB2 contribute to the development of FSGS through a previously unknown mechanism involving SD-related proteins.
-
Melanocortin Therapy Ameliorates Podocytopathy and Proteinuria in Experimental Focal Segmental Glomerulosclerosis Involving a Podocyte Specific Non-Mc1r-mediated Melanocortinergic Signaling.
Yingjin Qiao,Pei Wang,Mingyang Chang,Bohan Chen,Yan Ge,Deepak K. Malhotra,Lance D. Dworkin,Rujun Gong
DOI: https://doi.org/10.1042/cs20200016
2020-01-01
Clinical Science
Abstract:The clinical effectiveness of adrenocorticotropin in inducing remission of steroid-resistant nephrotic syndrome points to a steroidogenic-independent anti-proteinuric activity of melanocortins. However, which melanocortin receptors (MCR) convey this beneficial effect and if systemic or podocyte-specific mechanisms are involved remain uncertain. In vivo, wild-type (WT) mice developed heavy proteinuria and kidney dysfunction following Adriamycin insult, concomitant with focal segmental glomerulosclerosis (FSGS) and podocytopathy, marked by loss of podocin and synaptopodin, podocytopenia and extensive foot process effacement on electron microscopy. All these pathologic findings were prominently attenuated by NDP-MSH, a potent non-steroidogenic pan-MCR agonist. Surprisingly, MC1R deficiency in MC1R-null mice barely affected the severity of Adriamycin-elicited injury. Moreover, the beneficial effect of NDP-MSH was completely preserved in MC1R-null mice, suggesting that MC1R is likely non-essential for the protective action. A direct podocyte effect seems to contribute to the beneficial effect of NDP-MSH, because Adriamycin-inflicted cytopathic signs in primary podocytes prepared from WT mice were all mitigated by NDP-MSH, including apoptosis, loss of podocyte markers, de novo expression of the podocyte injury marker desmin, actin cytoskeleton derangement and podocyte hypermotility. Consistent with in vivo findings, the podoprotective activity of NDP-MSH was fully preserved in MC1R-null podocytes. Mechanistically, MC1R expression was predominantly distributed to glomerular endothelial cells in glomeruli but negligibly noted in podocytes in vivo and in vitro, suggesting that MC1R signaling is unlikely involved in direct podocyte protection. Ergo, melanocortin therapy protects against podocyte injury and ameliorates proteinuria and glomerulopathy in experimental FSGS, at least in part, via a podocyte-specific non-MC1R-mediated melanocortinergic signaling.
-
Therapeutic Mechanism of Glucocorticoids on Cellular Crescent Formation in Patients With Antiglomerular Basement Membrane Disease.
Xiaomei Wu,Mingchao Zhang,Xiao Huang,Lihua Zhang,Caihong Zeng,Jiong Zhang,Zhihong Liu,Zheng Tang
DOI: https://doi.org/10.1016/j.amjms.2017.04.015
IF: 3.462
2017-01-01
The American Journal of the Medical Sciences
Abstract:Background: This study aimed to explore the therapeutic mechanism of glucocorticoids (GCs) in antiglomerular basement membrane disease. Materials and Mehods: Thirty-four patients with biopsy-proven antiglomerular basement membrane nephritis were divided into the following 2 groups: group 1 (patients treated with GCs, n = 22) and group 2 (patients who were not treated with GCs, n = 12). The expression of parietal epithelial cells (PECs), activated PECs and glucocorticoid receptors (GRs) was examined quantitatively and compared between the 2 groups. Correlations between GR expression in glomeruli and patients' clinicopathological indices were also analyzed. Resuls: Compared with patients in group 2, patients in group 1 showed lower levels of serum creatinine (SCr) (P = 0.03), average cellular crescent percentage (P = 0.005) and macrophages infiltrating in renal interstitium (P = 0.03). PECs (P = 0.007) and activated PECs (P = 0.03) were strongly detected in the cellular components of classic crescents, and both were significantly reduced in group 1 compared to group 2. GR expression either in glomeruli (P = 0.01) or interstitium (P = 0.009) was lower in group 1 after GCs treatment than in group 2. Additionally, GR expression in glomeruli was strongly correlated with renal function (SCr: r = 0.45, P = 0.009; eGFR: r = -0.35, P = 0.046), the proportion of cellular crescents (r = 0.67, P < 0.001), PECs (r = 0.64, P < 0.001) and activated PECs (r = 0.72, P < 0.001), and the degree of interstitial (r = 0.50, P = 0.004) and glomerular (r = 0.49, P = 0.007) macrophage infiltration. Conclusions: GCs might exert their therapeutic effects via inhibiting the activation and proliferation of PECs, as well as macrophage infiltration, which could contribute to crescent formation and determine renal survival. GRs are involved in this process as well.
-
Dissection of Glomerular Transcriptional Profile in Patients with Diabetic Nephropathy: SRGAP2a Protects Podocyte Structure and Function.
Yu Pan,Song Jiang,Qing Hou,Dandan Qiu,Jingsong Shi,Ling Wang,Zhaohong Chen,Mingchao Zhang,Aiping Duan,Weisong Qin,Ke Zen,Zhihong Liu
DOI: https://doi.org/10.2337/db17-0755
IF: 7.7
2018-01-01
Diabetes
Abstract:Podocytes play a pivotal role in maintaining glomerular filtration function through their interdigitated foot processes. However, the mechanisms that govern the podocyte cytoskeletal rearrangement remain unclear. Through analyzing the transcriptional profile of renal biopsy specimens from patients with diabetic nephropathy (DN) and control donors, we identify SLIT-ROBO rGTPase-activating protein 2a (SRGAP2a) as one of the main hub genes strongly associated with proteinuria and glomerular filtration in type 2 DN. Immunofluorescence staining and Western blot analysis revealed that human and mouse SRGAP2a is primarily localized at podocytes and largely colocalized with synaptopodin. Moreover, podocyte SRGAP2a is downregulated in patients with DN and db/db mice at both the mRNA and the protein level. SRGAP2a reduction is observed in cultured podocytes treated with tumor growth factor-beta or high concentrations of glucose. Functional and mechanistic studies show that SRGAP2a suppresses podocytemotility through inactivating RhoA/Cdc42 but not Rac1. The protective role of SRGAP2a in podocyte function also is confirmed in zebrafish, in which knockdown of SRGAP2a, a SRGAP2 ortholog in zebrafish, recapitulates podocyte foot process effacement. Finally, increasing podocyte SRGAP2a levels in db/db mice through administration of adenovirus-expressing SRGAP2a significantly mitigates podocyte injury and proteinuria. The results demonstrate that SRGAP2a protects podocytes by suppressing podocyte migration.
-
[Analysis on Association of Glucocorticoid Receptor Gene Polymorphism with Steroid-Resistance in Idiopathic Nephrotic Syndrome of Children].
Jian-wei Ye,Jie Ding,Jian-ping Huang,Yan Chen,Yong Yao,Hui-jie Xiao,Ji-yun Yang,Ying Shen,Qun Meng
DOI: https://doi.org/10.3760/j.issn:0578-1310.2003.09.007
2003-01-01
Abstract:OBJECTIVEThe nephrotic syndrome is defined by heavy proteinuria, edema, hypoalbuminemia, and hyperlipidemia. Idiopathic nephrotic syndrome (INS) mainly occurs in children, which is generally treated with glucocorticoids. The majority of patients are steroid-sensitive (SSINS) while steroid-resistance occurs in a subset of NS children (SRINS). Although intensive efforts have been undertaken to study the associations between SRINS and renal pathological changes, pharmacokinetics, and the GR density and binding affinity, the mechanisms underlying steroid-resistance are still not elucidated entirely. The authors hypothesized that it might be associated with polymorphisms in the glucocorticoid receptor gene (NR3C1). The study aimed to screen the NR3C1 gene for polymorphisms in genomic DNA samples from SRINS, SSINS children and control group, and to analyze the association of the polymorphisms in the NR3C1 gene and SRINS of children.METHODSTotally 39 SRINS and 67 SSINS children (81 males and 25 females with the mean age of 7 years) were involved in the study. Umbilical cord blood of 62 normal neonates and peripheral blood of 2 healthy volunteers were selected as controls. Genomic DNA was isolated from peripheral blood lymphocytes of all subjects. All the NR3C1-coding exons and intron-flanking portions were amplified by polymerase chain reaction (PCR). For polymorphism screen, PCR products were analyzed by denaturing high performance liquid chromatography (DHPLC). DNA fragments with aberrant elution profiles were re-amplified and sequenced directly.RESULTSTwelve aberrant elution profiles were identified with DHPLC in SRINS, SSINS and controls. Among them, 6 previously reported polymorphisms and 6 novel polymorphisms were confirmed by sequencing (198G > A, 200G > A, IVSD-16G > T, 1896C > T, 2166C > T, 2430T > C; novel, 1206C > T, 1374A > G, IVSG-68_IVSG-63delAAAAAA, 2193T > G, IVSH-9C > G, 2382C > T), and 3 groups of SNPs were in complete linkage disequilibrium, which resulted in 3 different haplotypes ([198G > A + 200G > A], [1374A > G + IVSG-68_IVSG-63delAAAAAA + IVSH-9C > G + 2382C > T], [1896C > T + 2166C > T + 2430T > C]). The last two genotypes were first reported. The genotype frequencies of the 2 novel haplotypes were 10.3% vs 1.5% in SRINS and SSINS, and 15.4% vs 7.5% in SRINS and SSINS, respectively. Other polymorphisms were relatively rare detectable both in patients and controls.CONCLUSIONTwelve polymorphisms in the NR3C1 gene were detected with the technique of DHPLC, of which six polymorphisms were identified at the first time. Two types of newly found haplotypes were associated with steroid-resistant idiopathic nephrotic syndrome of children, which might be responsible for steroid-resistance in partial idiopathic nephrotic syndrome of children.
-
The redox sensitive glycogen synthase kinase 3β suppresses the self-protective antioxidant response in podocytes upon oxidative glomerular injury.
Changbin Li,Yan Ge,Ai Peng,Rujun Gong
DOI: https://doi.org/10.18632/oncotarget.6303
2015-01-01
Oncotarget
Abstract:The redox sensitive glycogen synthase kinase (GSK) 3 has been recently implicated in the pathogenesis of proteinuric glomerulopathy. However, prior studies are less conclusive because they relied solely on chemical inhibitors of GSK3, which provide poor discrimination between the isoforms of GSK3 apart from potential off target activities. In murine kidneys, the beta rather than the a isoform of GSK3 was predominantly expressed in glomeruli and distributed intensely in podocytes. By employing the doxycycline-activated Cre-loxP site specific gene targeting system, GSK3 beta was successfully knocked out (KO) selectively in podocytes in adult mice, resulting in a phenotype no different from control littermates. Electron microscopy of glomeruli in KO mice demonstrated more glycogen accumulation in podocytes but otherwise normal ultrastructures. Upon oxidative glomerular injury induced by protein overload, KO mice excreted significantly less albuminuria and had much attenuated podocytopathy and glomerular damage. The anti-proteinuric and glomerular protective effect was concomitant with diminished accumulation of reactive oxygen species in glomeruli in KO mice, which was likely secondary to a reinforced Nrf2 antioxidant response in podocytes. Collectively, our data suggest that GSK3 beta is dispensable for glomerular function and histology under normal circumstances but may serve as a therapeutic target for protecting from oxidative glomerular injuries.
-
GENETICS IN ENDOCRINOLOGY: Glucocorticoid resistance syndrome
Géraldine Vitellius,Marc Lombes
DOI: https://doi.org/10.1530/EJE-19-0811
2020-02-01
Abstract:Glucocorticoids (GC) such as cortisol regulate multiple physiological functions, notably those involved in development, metabolism, inflammatory processes and stress, and exert their effects upon binding to the glucocorticoid receptor (GR, encoded by NR3C1 gene in humans). GC signaling follows several consecutive steps leading to target gene transactivation, including ligand binding, nuclear translocation of ligand-activated GR complexes, DNA binding, and recruitment of functional transcriptional machinery. Generalized glucocorticoid resistance syndrome, due to GR loss-of-function mutations, may be related to the impairment of one of the GC signaling steps. To date, 31 NR3C1 loss-of-function mutations have been reported in patients presenting with various clinical signs such as hypertension, adrenal hyperplasia, hirsutism or metabolic disorders associated with biological hypercortisolism but without Cushing syndrome signs and no negative regulatory feedback loop on the hypothalamic-pituitary-adrenal axis. Functional characterization of GR loss-of-function mutations often demonstrates GR haploinsufficiency and a decrease of GR target gene induction in relevant cell types. The main signs at presentation are very variable from resistant hypertension, bilateral adrenal hyperplasia likely related to increased ACTH levels but not exclusively, hirsutism to isolated renin-angiotensin-aldosterone system abnormalities in a context of 11βHSD2 deficiency. Some mutated GR patients are obese or overweight together with a healthier metabolic profile that remains to be further explored in future studies. Deciphering the molecular mechanisms altered by GR mutations should enhance our knowledge on GR signaling and ultimately facilitate management of GC-resistant patients. This review also focuses on the criteria facilitating identification of novel NR3C1 mutations in selected patients.
-
Expression of GRα, GRβ in Peripheral Blood Mononuclear Cells in Patients with Primary Nephrotic Syndrome and Its Clinical Significance
QIU Lin,FENG Qi-hua,ZHAO Hui-jun
DOI: https://doi.org/10.3969/j.issn.1000-3606.2012.04.010
2012-01-01
Abstract:Objective To investigate the expression of glucocorticoid receptor(GR)α,GRβ in peripheral blood mononuclear cells in patients with primary nephrotic syndrome and the possible mechanism of GRα and GRβ mediated the steroid resistance.Methods Fifteen children with glucocorticoid-sensitive primary nephritic syndrome(SSNS group),15 children with glucocorticoid-resistant primary nephritic syndrome(SRNS group),and 10 healthy children(control group)were recruited.The expression of GRα and GRβ mRNA in peripheral blood mononuclear cells(PMBC)was detected by reverse transcription polymerase chain reaction.The relationship of the expression of GRα and GRβ with 24 h urinary protein and renal pathological score were analyzed.Results Both GRα and GRβ,mainly GRα,were expressed in PMBC in children with primary nephritic syndrome.The expression of GRα mRNA were not significant different among SRNS,SSNS and control groups(P 0.05).The expression of GRβ mRNA and GRα/GRβ mRNA was significantly higher in SRNS group than that in SSNS and control groups(P 0.05).The expression of GRβ was positively correlated with 24 h urinary protein and renal pathological score(both P 0.05).Conclusions The expression of GRβ mRNA can be used as an indicator to monitor the condition and prognosis of children with primary nephritic syndrome.Because the expression of GRβ is increased in SRNS patients,increased sensitivity of hormone receptor is expected to provide new ideas for the treatment of SRNS.
-
R168H and V165X Mutant Podocin Might Induce Different Degrees of Podocyte Injury Via Different Molecular Mechanisms
Qingfeng Fan,Han Zhang,Jie Ding,Shufang Liu,Jing Miao,Yan Xing,Zihua Yu,Na Guan
DOI: https://doi.org/10.1111/j.1365-2443.2009.01336.x
2009-01-01
Genes to Cells
Abstract:A lot of mutations of podocin, a key protein of podocyte slit diaphragm (SD), have been found both in hereditary and sporadic focal segmental glomeruloscleorosis (FSGS). Nevertheless, the mechanisms of podocyte injury induced by mutant podocins are still unclear. A compound heterozygous podocin mutation was identified in our FSGS patient, leading to a truncated (podocin V165X) and a missense mutant protein (podocin R168H), respectively. Here, it was explored whether and how both mutant podocins induce podocyte injury in the in vitro cultured podocyte cell line. Our results showed that podocin R168H induced more significant podocyte apoptosis and expression changes in more podocyte molecules than podocin V165X. Podocyte injury caused by the normal localized podocin V165X was effectively inhibited by TRPC6 knockdown. The abnormal retention of podocin R168H in endoplasmic reticulum ( ER) resulted in the mis-localizations of other critical SD molecules nephrin, CD2AP and TRPC6, and significantly up-regulated ER stress markers Bip/grp78, p-PERK and caspase-12. These results implicated that podocin R168H and podocin V165X induced different degrees of podocyte injury, which might be resulted from different molecular mechanisms. Our findings provided some possible clues for further exploring the pharmacological targets to the proteinuria induced by different mutant podocins.
-
CRB2 Depletion Induces YAP Signaling and Disrupts Mechanosensing in Podocytes
Yingyu Sun,Nils M. Kronenberg,Sidharth K. Sethi,Surjya N. Dash,Maria E. Kovalik,Benjamin Sempowski,Shelby Strickland,Rupresh Raina,C. John Sperati,Xuefei Tian,Shuta Ishibe,Gentzon Hall,Malte C Gather
DOI: https://doi.org/10.1101/2024.10.22.619513
2024-10-26
Abstract:Focal Segmental Glomerulosclerosis (FSGS) is a histologic lesion caused by a variety of injurious stimuli that lead to dysfunction/loss of glomerular visceral epithelial cells (i.e. podocytes). Pathogenic mutations in CRB2, encoding the type 1 transmembrane protein Crumb 2 Homolog Protein, have been shown to cause early-onset corticosteroid-resistant nephrotic syndrome (SRNS)/FSGS. Here, we identified a 2-generation East Asian kindred (DUK40595) with biopsy-proven SRNS/FSGS caused by a compound heterozygous mutation in CRB2 comprised of the previously described truncating mutation p.Gly1036_Alafs*43 and a rare 9-bp deletion mutation p.Leu1074_Asp1076del. Because compound heterozygous mutations involving the truncating p.Gly1036_Alafs*43 variant have been associated with reduced CRB2 expression in podocytes and autosomal recessive SRNS/FSGS, we sought to define the pathogenic effects of CRB2 deficiency in podocytes. We show that CRB2 knockdown induces YAP activity and target gene expression in podocytes. It upregulates YAP-mediated mechanosignaling and increases the density of focal adhesion and F-actin. Using Elastic Resonator Interference Stress Microscopy (ERISM), we demonstrate that CRB2 knockdown also enhances podocyte contractility in a substrate stiffness-dependent manner. The knockdown effect decreases with increasing substrate stiffness, indicating impaired mechanosensing in CRB2 knockdown cells at low substrate stiffness. While the mechanical activation of CRB2 knockdown cells is associated with increased YAP activity, the enhanced cell contractility is not significantly reduced by the selective YAP inhibitors K-975 and verteporfin, suggesting that multiple pathways may be involved in mechanosignaling downstream of CRB2. Taken together, these studies provide the first evidence that CRB2 deficiency may impair podocyte mechanotransduction via disruption of YAP signaling in podocytes.
Biophysics
-
Comparative Proteomic Analysis of Children FSGS FFPE Tissues.
Jiajia Ni,Sha Tian,Lin Bai,Qianying Lv,Jialu Liu,Jiaojiao Liu,Ye Fang,Yihui Zhai,Qian Shen,Jia Rao,Chen Ding,Hong Xu
DOI: https://doi.org/10.1186/s12887-022-03764-7
2022-01-01
BMC Pediatrics
Abstract:BACKGROUND:In children, focal segmental glomerulosclerosis (FSGS) is the main cause of steroid resistant nephrotic syndrome (SRNS). To identify specific candidates and the mechanism of steroid resistance, we examined the formalin-fixed paraffin embedded (FFPE) renal tissue protein profiles via liquid chromatography tandem mass spectrometry (LC-MS/MS).METHODS:Renal biopsies from seven steroid-sensitive (SS) and eleven steroid-resistant (SR) children FSGS patients were obtained. We examined the formalin-fixed paraffin embedded (FFPE) renal tissue protein profiles via liquid chromatography tandem mass spectrometry (LC-MS/MS). Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment and Gene Ontology (GO) analysis, as well as the construction of protein-protein interaction (PPI) network were performed. Two proteins were further valiadated by immunohistochemistry staining in FSGS patients and mice models.RESULTS:In total, we quantified more than 4000 proteins, of which 325 were found to be differentially expressed proteins (DEPs) between the SS and SR group (foldchange ≥2, P<0.05). The results of GO revealed that the most significant up-regulated proteins were primarily related to protein transportation, regulation of the complement activation process and cytolysis. Moreover, clustering analysis showed differences in the pathways (lysosome, terminal pathway of complement) between the two groups. Among these potential candidates, validation analyses for LAMP1 and ACSL4 were conducted. LAMP1 was observed to have a higher expression in glomerulus, while ACSL4 was expressed more in tubular epithelial cells.CONCLUSIONS:In this study, the potential mechanism and candidates related to steroid resistance in children FSGS patients were identified. It could be helpful in identifying potential therapeutic targets and predicting outcomes with these proteomic changes for children FSGS patients.
-
An overview of molecular mechanism of nephrotic syndrome
Juliana Reis Machado,Laura Penna Rocha,Precil Diego Miranda de Menezes Neves,Eliângela de Castro Cobô,Marcos Vinícius Silva,Lúcio Roberto Castellano,Rosana Rosa Miranda Corrêa,Marlene Antônia Reis
DOI: https://doi.org/10.1155/2012/937623
Abstract:Podocytopathies (minimal change disease (MCD) and focal segmental glomerulosclerosis (FSGS)) together with membranous nephropathy are the main causes of nephrotic syndrome. Some changes on the expression of nephrin, podocin, TGF-β, and slit diaphragm components as well as transcription factors and transmembrane proteins have been demonstrated in podocytopathies. Considering the pathogenesis of proteinuria, some elucidations have been directed towards the involvement of epithelial-mesenchymal transition. Moreover, the usefulness of some markers such as TGF-β1, nephrin, synaptopodin, dystroglycans, and malondialdehyde have been determined in the differentiation between MCD and FSGS. Experimental models and human samples indicated an essential role of autoantibodies in membranous glomerulonephritis, kidney damage, and proteinuria events. Megalin and phospholipase-A2-receptor have been described as antigens responsible for the formation of the subepithelial immune complexes and renal disease occurrence. In addition, the complement system seems to play a key role in basal membrane damage and in the development of proteinuria in membranous nephropathy. This paper focuses on the common molecular changes involved in the development of nephrotic proteinuria.
-
Podocin and Beta Dystroglycan expression to study Podocyte-Podocyte and basement membrane matrix connections in adult protienuric states
Praveen B Shankar,Ritambhra Nada,Kusum Joshi,Ashwani Kumar,Charan Singh Rayat,Vinay Sakhuja
DOI: https://doi.org/10.1186/1746-1596-9-40
2014-02-21
Diagnostic Pathology
Abstract:BackgroundPodocytes can be the primary site of injury or secondarily involved in various protienuric states. Cross talk between adjacent foot processes and with basement membrane is important for slit diaphragm function. Does expression of podocyte associated proteins in kidney biopsies alter with site/type of primary injury? Genetic mutations of podocin result in steroid resistant FSGS. Can protein expression of podocin predict resistant cases to initiate further genetic evaluation?MethodsAdult patients (n-88) with protienuria- minimal change disease(MCD)-22, focal segmental glomerulosclerosis(FSGS)-21,membranous glomerulonephritis(MGN)-25 and IgA nephropathy(IgAN)-20 were selected for immunohistochemistry with podocin and beta dystroglycan . Results were graded (0 - 3+scale )and compared with control biopsies and internal control. Treatment and follow up (6 months -2 ½ years) of FSGS and MCD cases were collected.ResultsThere was intense to moderate staining of the podocytes with podocin and β dystroglycan in the glomeruli in all cases (MCD, FSGS, IgAN and MGN) except for weak staining with β dystroglycan in 3 cases of MCD. There was loss of immunostains in areas of segmental/global sclerosis. There was no significant difference in the staining pattern between the groups. In primary podocytopathies, staining pattern did not differ between steroid resistant, sensitive or dependent cases.ConclusionsImmunohistochemical expression of podocin and β dystroglycan does not differ in nephropathies which have different site of injury depending on absence (MCD and FSGS) or presence of immune deposits and their localization (MGN and IgAN). Podocin and β dystroglycan staining did not differentiate steroid sensitive and resistant cases, hence, does not give clue to initiate genetic studies. However, analysis of bigger cohort may be required.SummaryPodocin and β dystroglycan immunohistochemistry was done to analyze podocyte - podocyte and podocyte -basement membrane matrix connections in adult protienuric states. Primary podocytopathies i.e. MCD and FSGS and secondary podocytopathy due to immune complex deposition i.e. MGN (subepithelial) and IgAN (mesangial) were analyzed. There was no difference in staining patterns between primary and secondary podocytopathies or between steroid sensitive, resistant and dependent cases of FSGS and MCD.Virtual slidesThe virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/2258608781052786
pathology
-
Blocking Ribosomal Protein S6 Phosphorylation Inhibits Podocyte Hypertrophy and Focal Segmental Glomerulosclerosis.
Fang Li,Yili Fang,Qiyuan Zhuang,Meichu Cheng,Desmond Moronge,Hao Jue,Oded Meyuhas,Xiaoqiang Ding,Zhigang Zhang,Jian-Kang Chen,Huijuan Wu
DOI: https://doi.org/10.1016/j.kint.2022.02.037
IF: 19.6
2022-01-01
Kidney International
Abstract:Ribosomal protein S6 (rpS6) phosphorylation mediates the hypertrophic growth of kidney proximal tubule cells. However, the role of rpS6 phosphorylation in podocyte hypertrophy and podocyte loss during the pathogenesis of focal segmental glomerulosclerosis (FSGS) remains undefined. Here, we examined rpS6 phosphorylation levels in kidney biopsy specimens from patients with FSGS and in podocytes from mouse kidneys with Adriamycin-induced FSGS. Using genetic and pharmacologic approaches in the mouse model of FSGS, we investigated the role of rpS6 phosphorylation in podocyte hypertrophy and loss during development and progression of FSGS. Phosphorylated rpS6 was found to be markedly increased in the podocytes of patients with FSGS and Adriamycin-induced FSGS mice. Genetic deletion of the Tuberous sclerosis 1 gene in kidney glomerular podocytes activated mammalian target of rapamycin complex 1 signaling to rpS6 phosphorylation, resulting in podocyte hypertrophy and pathologic features similar to those of patients with FSGS including podocyte loss, leading to segmental glomerulosclerosis. Since protein phosphatase 1 is known to negatively regulate rpS6 phosphorylation, treatment with an inhibitor increased phospho-rpS6 levels, promoted podocyte hypertrophy and exacerbated formation of FSGS lesions. Importantly, blocking rpS6 phosphorylation (either by generating congenic rpS6 knock-in mice expressing non-phosphorylatable rpS6 or by inhibiting ribosomal protein S6 kinase 1-mediated rpS6 phosphorylation with an inhibitor) significantly blunted podocyte hypertrophy, inhibited podocyte loss, and attenuated formation of FSGS lesions. Thus, our study provides genetic and pharmacologic evidence indicating that specifically targeting rpS6 phosphorylation can attenuate the development of FSGS lesions by inhibiting podocyte hypertrophy and associated podocyte depletion.