Skip to main content
PLOS One logoLink to PLOS One
. 2025 Mar 10;20(3):e0319424. doi: 10.1371/journal.pone.0319424

Podocyte specific knockout of the natriuretic peptide clearance receptor is podocyte protective in focal segmental glomerulosclerosis

Liming Wang 1, Yuping Tang 1, Anne F Buckley 2, Robert F Spurney 1,*
Editor: Michael Bader3
PMCID: PMC11892885  PMID: 40063586

Abstract

Natriuretic peptides (NPs) bind to glomerular podocytes and attenuate glomerular injury. The beneficial effects of NPs are negatively regulated by the NP clearance receptor (NPRC), which is highly expressed in podocytes. To determine if inhibiting NPRC is podocyte protective, we examined the effects of deleting NPRC in both cultured podocytes and in vivo. We found that: 1.Both atrial NP and C-type NP inhibit podocyte apoptosis in cultured podocytes, but these podocyte protective effects are significantly attenuated in cells expressing NPRC, and 2. Atrial NP was significantly more effective than CNP at inhibiting the apoptotic response. Consistent with the protective actions of NPs, podocyte specific knockout of NPRC reduced albuminuria, glomerular sclerosis and tubulointerstitial inflammation in a mouse model of focal segmental glomerulosclerosis. These beneficial actions were associated with: 1. Decreased expression of the myofibroblast marker alpha-smooth muscle actin, 2. Reduced expression of the extracellular matrix proteins collagen 4-alpha-1 and fibronectin, and 3. Preserved expression of the podocyte proteins nephrin and podocin. Inhibiting NP clearance may be a useful therapeutic approach to treat glomerular diseases.

Introduction

Focal segmental glomerulosclerosis (FSGS) is an important cause of kidney disease worldwide [1]. The disease is characterized by heavy proteinuria, scarring of the glomerulus in a segmental fashion and frequent progression to end stage kidney disease (ESKD [2]. Treatment is directed at controlling hypertension and reducing proteinuria by blockade of the renin-angiotensin system [3]. More recently, sodium glucose transporter 2 inhibitors (SGLT2i) have further slowed disease progression [46]. Despite these advances, nephrotic patients with FSGS remain at significant risk for the development of ESKD [3].

Glomerular podocytes are terminally differentiated cells that play a key role in maintaining both the structure of the filtering apparatus and glomerular permselectivity [2,7,8]. While multiple mechanisms cause FSGS, the unifying feature is podocyte injury that leads to a reduction in the number of viable podocytes [2,7]. Because podocytes are unable to proliferate, the decrease in glomerular podocytes promotes damage to the filtering apparatus, which eventually causes glomerulosclerosis (GS), and renal failure.

NPs bind to cell surface NP receptors (NPRs) [911]. Atrial NP (ANP) and brain NP (BNP) bind NPR type A (NPRA, also termed Guanylyl Cyclase-A or GC-A), and CNP binds NPR type B (NPRB, also termed Guanylyl Cyclase-B or GC-B) [911]. The NP clearance receptor (NPRC) inhibits the effects of NPs by binding and degrading ANP, BNP and CNP [1013]. The negative regulatory actions of NPRC play an important role at inhibiting the effects of NPs in podocytes because: 1. Podocytes express NPRA, NPRB and NPRC [12,1417], and 2. NPRC is highly expressed in glomerular podocytes compared to other glomerular and kidney cell types [12,1417].

Recent studies suggest that NPs have beneficial actions in glomerular diseases [15,1722]. For example, KO of the cGMP generating ANP/BNP receptor (NPRA) exacerbates glomerular injury in proteinuric mouse models [15,22]. Moreover, TG overexpression of BNP ameliorates diabetic kidney disease [18] and immune medicated renal injury [21] in mouse models. These beneficial effects of NPs are mediated, at least in part, by stimulating cGMP generation [9,20]. This increase in cGMP generation inhibits signaling pathways that play important roles in kidney diseases including calcium signaling, Rho GTPases and TGF-beta [9,20].

We hypothesized that inhibition of NP clearance by NPRC would increase the local concentration of NPs, increase cGMP signaling in glomerular podocytes and inhibit glomerular injury. In support of this hypothesis, we previously found that: 1. NPs potently stimulated cGMP generation and inhibited podocyte apoptosis in cultured podocytes [17], and 2. Pharmacologic blockade of NRPC in vivo enhanced urinary cGMP excretion and significantly reduced albuminuria in a mouse model of FSGS [17]. To further test the hypothesis in vivo, we deleted NPRC [9] specifically in glomerular podocytes in a transgenic (TG) mouse model of FSGS created in our laboratory [23], This model sensitizes mice to the podocyte toxin puromycin aminonucleoside (PAN). In these TG mice, a single dose of PAN induces GS and heavy proteinuria. We found that podocyte specific NPRC KO significantly reduced albuminuria, GS and TI inflammation, inhibited both myofibroblast activation and deposition of fibronectin and collagen 4 (alpha-1 chain) in glomeruli, and preserved expression of the podocyte proteins nephrin and podocin. These data suggest that inhibition of NP clearance by NPRC may be a useful therapeutic approach to treat FSGS and perhaps other glomerular diseases.

Results

Podocyte protective effects of NPs in cultured podocytes

We previously found that both ANP and CNP protected cultured podocytes from apoptotic stimuli [17]. ANP is mainly produced by the heart and acts in an endocrine fashion to affect the function of target organs [24]. In contrast, CNP is produced and acts locally in an autocrin and paracrine fashion [24]. There is strong evidence that that ANP has potent podocyte protective actions in animal models of glomular disease [15,1719,21,22], but little is known about the paracrine/endocrine actions of CNP in the kidney.

CNP is expressed in both the tubules and glomeruli by in-situ hybridization [25]. In glomeruli, CNP was detected in both the parietal and visceral layers of the glomerulus [25]. The presence of CNP mRNA in the viseral layer suggests that CNP is expressed in podocytes (visceral epithelial cells). Consistent with these findings, CNP was detecteded in cultured mouse podocytes by RT-PCR [12]. Moreover, the CNP receptor (NPRB) is expressed by podocytes [12,14,16,17] and stimulates cGMP generation [12,17]. Taken together, these data indicate that CNP production by glomerular podocytes might act in an autocrine, podocyte protective fashion as depicted in Figs 1A and 1B.

Fig 1. (1A) CNP is produced by podocytes and binds to both NPRB and NPRC on glomerular podocytes.

Fig 1

(1B) Knockdown (KD) of NPRC increases CNP levels by inhibiting clearance of CNP from the circulation. (1C) Four shRNA constructs (1, 2, 3 & 4) and a scrambled control were used to KD NPRC in cultured podocytes. Constructs 1 and 2 provided the most effective KD of NPRC. (1D & 1E) CNP expression in cell culture medium was significantly increased by KD of NPRC in the both the presence and absence of the neprilysin inhibitor LBQ657. (1F) Generation of cGMP by low dosages of CNP was enhanced by NPRC KD in cultured podocytes compared to controls. (1G) Serum deprivation significantly increased apoptosis in control podocytes and KD of NPRC inhibited the apoptotic response in the absence of exogenous NPs. In addition, treatment with the NPRB antagonist P19 inhibited the podocyte protective actions of NPRC KD, resulting in an increase in apoptosis. * P <  0.005 **P <  0.001 versus control cells.

We first determined if inhibiting CNP clearance by knockdown (KD) of NPRC increased the autocine podocyte protective effects of CNP. For the studies, we used 4 shRNA constructs (1, 2, 3 & 4) and a scrambled control construct to KD NPRC. As shown in Fig 1C, constructs 1 & 2 provided the most effective KD of NPRC after selection, and these cell lines were chosen for the apoptosis studies. To further confirm KD, we examined the effect of low dose CNP on cGMP generation in cultured mouse podocytes. Given that podocytes express high levels of neprilysin [26,27], we measured CNP in the presence or absence of the neprilysin inhibitor LBQ657 [28]. As shown in Fig 1D and 1E, there was a significant increase in CNP levels in NPRC KD cells compared to wild type (WT) podocytes in both the presence and absence of LBQ657. We also examined the effect of NPRC KD on NPRB signaling by measuring cGMP generation (Fig 1F). In KD cells, cGMP generation was significantly increased compared to control cells. Lastly, we evaluated: 1. The effects of serum deprivation on apoptosis in NPRC-KD cells and controls, and 2. The effect of pharmacologic blockade of the CNP receptor (NPRB) on the apoptotic response. As shown in Fig 1G, serum deprivation significantly increased apoptosis in control cells, and the apoptotic response was significantly reduced in the KD podocytes in the absence of adding exogenous NPs to the culture medium. Moreover, pharmacologic blockade of NPRB with P19 [29,30] attenuated the podocyte protective effect in NPRC KD cells. Lastly, similar beneficial effects of NPRC KD on apoptosis were observed using a shorter duration of serum deprivation (S1 Fig in S1 Data).

We next examined expression of NPRs in cultured podocytes and compared the podocyte protective effects of both ANP and CNP. Fig 2A shows NPRA, NPRB and NPRC mRNA expression in cultured podocytes. KD of NPRC effectively reduced NPRC mRNA in cultured podocytes (P < 0.0001). In addition, NPRC was highly expressed in WT podocytes compared to either NPRA or NPRB in the presence of absence of NPRC KD (P < 0.0001). Fig 2B demonstrates the differences in NPRA and NPRB expression by enlarging the vertical axis (ordinate) of Fig 2A. The mRNA levels of NPRA and NPRB were similar in cells expressing NPRC, and NPRA expression levels were not significantly changed by NPRC KD. NPRB was, however, significantly decreased in NPRC KD cells. Fig 2C shows the effects of 10 nM ANP and 10 nM CNP on cGMP generation. There was a stepwise increase in cGMP generation in both WT and KD podocytes. At the 10 nM concentrations, the increase in cGMP generation was statistically significant in the KD podocytes stimulated with either ANP or CNP compared to unstimulated KD podocytes and compared to WT podocytes expressing NPRC. In addition, we investigated the relative podocyte protective actions of ANP and CNP in cultured podocytes. As shown in Fig 2D, serum deprivation increased podocyte apoptosis and KD of NPRC reduced the apoptotic response in the absence of additional NPs added to the culture medium, similar to Fig 1G. Treatment with 10 nM ANP was, however, more effective at decreasing apoptosis in NPRC KD cells compared NPRC WT cells expressing NPRC. Moreover, combined ANP treatment with NPRC KD was the most effective approach to reduce apoptotic response. In contrast. treatment with 10 nM CNP alone had no significant effect on the apoptotic response in podocytes expressing NPRC, although NPRC KD inhibited podocyte apoptosis with or without the addition of 10 nM CNP. Taken together, these data suggest that ANP is more effective than CNP at inhibiting the apoptotic response and combining NPRC KD with ANP treatment further augmented the podocyte protective effect.

Fig 2. (A) NPRC was highly expressed in WT podocytes compared to either NPRA or NPRB in the presence of absence of NPRC KD.

Fig 2

In addition, NPRC KD effectively decreased NPRC mRNA in cultured podocytes. (B) NPRB expression was downregulated in NPRC KD cells. (C) Both ANP and CNP increased cGMP generation in a stepwise fashion. The increase in cGMP generation was statistically significant for the KD cells at the 10 nM concentrations compared to both unstimulated KD cells and WT podocytes stimulated with either ANP or CNP. (D) Apoptosis induced by serum deprivation was inhibited by KD of NPRC. ANP treatment alone further inhibited podocyte apoptosis and combined treatment with ANP and NPRC KD reduced the apoptotic response toward baseline. In contrast, CNP alone had little effect on podocyte apoptosis, but NPRC KD reduced the apoptotic response in the presence or absence of 10 nM CNP. **P < 0.0001 versus other NPRs shown in Fig 2A.

Podocyte specific KO of NPRC in a mouse model of FSGS

To determine if inhibition of NP clearance had beneficial effects in a proteinuric kidney disease, we deleted NPRC specifically in glomerular podocytes in a TG mouse model of FSGS created in our laboratory [23]. As described in the Methods Section, these “double” TG mice express a doxycycline inducible, constitutively active Gq alpha-subunit (GqQ > L) specifically in podocytes [23]. Following induction of GqQ > L, a single dose of PAN causes robust albuminuria in Gq mice, but only mild disease in WT mice. To knockout NPRC in podocytes, Gq mice were crossed with TG mice expressing: 1. Two NPRC “floxed” alleles flanking exon 3 of NPRC [17], and 2. Cre-recombinase under the control of a doxycycline responsive promoter (see Methods). This created “triple” transgenic mice with 2 “floxed” NPRC alleles. In these animals, doxycycline induces podocyte specific GqQ > L expression and podocyte specific KO of NPRC on demand (Gq-KO mice). Control mice did not express GqQ > L and included WT NPRC KO mice (WT-KO mice) and WT mice expressing NPRC (WT mice).

We first determined the efficiency of podocyte specific NPRC-KO by crossing Gq mice with a reporter mouse expressing a two-color fluorescent Cre reporter allele [31]. The reporter allele contains a cell membrane-localized red fluorescence protein (tdTomato) with widespread expression in all tissues and cell types prior to induction of Cre recombinase. Expression of Cre recombinase induces a cell membrane-localized green fluorescence (EGFP) in cells expressing Cre recombinase, with red fluorescence confined to other cell types. As shown in Fig 3A, green fluorescence was induced in glomeruli of tdTomato mice following induction of Cre recombinase by doxycycline. Little EGFP expression was observed in mice expressing only the reporter construct (Fig 3B). A low power view of the tdTomato studies is shown Supplementary S2 Fig in S1 Data.

Fig 3. Cre-mediated recombination in vivo.

Fig 3

(3A & 3B). These studies used a mouse expressing a reporter allele that contains a cell membrane-localized red fluorescence transgene (td-Tomato) with widespread expression in all tissues and cell types. Expression of Cre recombinase induces a cell membrane-localized green fluorescence protein (EGFP). To examine Cre-medicated recombination in vivo, “double” TG mice (NPHS2-rtTA, tetO-Cre) mice were crossed with the TG reporter animals to create triple TG mice (NPHS2-rtTA, tetO-Cre, td-Tomato). In these TG mice, treatment with doxycycline induces green fluorescence specifically in podocytes, with red fluorescence confined to other cell types (panel 3A). In contrast, little EGFP expression was observed in controls (panel 3B). (3C) Quantitative RT-PCR demonstrated a significant decrease in expression NPRC in enriched glomerular preparations from NPRC KO mice compared to controls. (3D & 3E). NPRC protein was significantly reduced in glomerular preparations form podocyte specific KO mice compared to WT mice. Expression of NPRC in kidney cortices was similar in WT and KO kidneys.

We also measured expression of NPRC mRNA and NPRC protein in enriched glomerular preparations from both WT mice and KO mice using quantitative RT-PCR and immunoblotting. As shown in Fig 3C, 3D and 3E, there was a significant decrease in both NPRC mRNA and NPRC protein in glomerular preparations from KO mice compared to WT mice. Expression of NPRC in kidney cortices was similar in WT and KO kidneys.

Systemic BP and albuminuria

We next evaluated the effects of NPRC-KO on systemic blood pressure (BP) and proteinuria using the protocol described in the Methods Section (see schematic in Supplementary S3 Fig). As shown in Fig 4A, systolic BP was similarly reduced in both groups of Gq mice compared to controls. Although there is variability in the BP results, the BP is similar in the Gq mice (GQ and Gq-KO), suggesting that a difference in BP did not affect the severity of renal injury in the Gq groups. Fig 4B shows the effects of treatment with PAN on proteinuria in Gq mice. PAN induced heavy proteinuria on days 10 and 14 in Gq-mice compared to baseline. Podocyte specific KO of NPRC significantly reduced albuminuria at both the 10-day and 14-day time points. PAN had little effect on albuminuria in controls (supplemental S1 Table in S1 Data).

Fig 4. Effect of podocyte specific NPRC KO on systemic BP and albuminuria.

Fig 4

(4A) Systolic BP was similarly decreased in both groups of Gq mice compared to controls, but this difference was not statistically significant. (4B) Treatment with PAN induced heavy proteinuria at day 10 and day 14 in WT Gq mice. The increase in albuminuria was significantly decreased by NPRC-KO at both day 10 and day 14 following PAN injection.

Renal histopathology

For these studies, histopathologic abnormalities were graded by a pathologist blinded to genotype using previously described criteria [32] (see Methods). Figs 5A and 5B show representative pictures of GS in Gq mice and Gq-KO mice. As shown in Fig 5C, there was a significant decrease in GS in Gq-KO mice compared to Gq mice. A similar significant decrease was observed for tubulointerstitial (TI) inflammation (Fig 5D). Lastly, tubular injury tended to be more severe in the WT Gq mice compared to Gq-KO mice (Fig 5E), but the overall difference was not statistically significant (Fig 5E). Representative low power pictures of the histopathology in the controls and Gq groups are shown in supplementary S4 & S5 Figs). In addition, the number of mice with mild, moderate and severe GS is reported in supplementary S6 Fig.

Fig 5. Effect of podocyte specific NPRC KO on renal histopathology.

Fig 5

(5A, 5B) Representative pictures of GS in Gq mice and Gq-KO mice. (5C) NPRC-KO significant reduced GS in Gq-KO mice compared to Gq mice. (5D).NPRC-KO significantly reduced TI inflammation in Gq-KO mice compared to Gq mice. (5E) Tubular injury was similar in both groups of Gq mice. (NOTE: data in Figs 5C, 5D and 5E are presented as a percentage of mice with the indicated histologic abnormalities, but the statistical evaluation was performed using the number of mice with the specified histologic abnormality).

Glomerular fibrosis

To further evaluate the effects of NPRC KO on glomerular fibrosis, we measured fibrotic markers in enriched glomerular preparations. For these studies, data from WT mice and WT-KO mice were similar and were combined for the analyses (WT controls). Figs 6A and 6B show expression of the myofibroblast differentiation marker alpha-smooth muscle actin (alpha-SMA) and the extracellular matrix protein (ECM) fibronectin. There was a statistically significant increase in glomerular expression of mRNA for alpha-SMA in Gq mice compared to WT controls, and KO of NPRC significantly reduced alpha-SMA in Gq-KO mice. A similar pattern was observed for fibronectin, but the decrease did not reach statistical significance. We also evaluated expression of the alpha-1 chains of collagen 4 (COL4 alpha-1) and collagen 1 (Col1 alpha-1) in the glomerular preparations (Figs 6C and 6D). Col4a1 is a major component of the FSGS lesion [3335], and type 1 collagens are significantly increased in both global GS [36,37] and TI fibrosis [34,38]. As shown in Fig 6C, there was a significant increase COL4 alpha-1 mRNA in glomerular preparations from Gq mice compared to WT controls, and this increase in COL4 alpha-1 in Gq mice was significantly inhibited in Gq KO mice. In contrast, there were no significant differences in expression of COL1 alpha-1 in the glomerular preparations (6D).

Fig 6. Effect of NPRC KO on fibrotic markers.

Fig 6

(6A) The myofibroblast marker alpha-SMA was significantly increased in glomerular preparations from WT Gq mice compared to controls. The increase in alpha-SMA was significantly reduced by podocyte specific KO of NPRC. (6B, 6C & 6D) The ECM proteins fibronectin and the alpha-1 chains of collagen 4 (CO4 alpha-1) and collagen 1 (Col1 alpha-1) were significantly increased in Gq mice compared to controls. Podocyte specific KO of NPRC decreased expression of both fibronectin and Col4 alpha-1, which was statistically significant for Col4 alpha-1. In contrast, NPRC KO had little effect on Col1 alpha-1 expression. (6E, 6F, 6G & 6H) Protein levels of the myofibroblast marker alpha-SMA and fibronectin were significantly increased in glomerular preparations from WT Gq mice compared to controls. Podocyte specific KO of NPRC significantly reduced expression of both alpha-SMA and the ECM protein fibronectin. NOTE: Quantitative RT-PCR data is presented in logarithmic format.

To confirm the mRNA studies, we evaluated expression of fibrotic markers including the myofibroblast marker alpha-SMA and the extracellular matrix protein fibronectin in glomerular preparations by immunoblotting. As shown in Figs 5E and 5F, expression alpha-SMA was significantly increased in glomerular preparations from Gq mice compared to WT controls, and this increase was significantly reduced by KO of NPRC. Similarly, fibronectin expression was significantly increased in glomerular preparations from Gq mice compared to WT controls and this increase in fibronectin deposition was significantly reduced by KO of NPRC (Figs 6G and 6H).

Podocyte protective effects of NPRC KO

In addition to fibrotic markers, we also examined the effect of podocyte specific KO of NPRC on the podocyte proteins nephrin and podocin. As shown in Figs 7A and 7B, expression of both nephrin and podocin mRNAs were significantly decreased in glomerular preparations from Gq mice, and podocyte specific KO of NPRC significantly increased nephrin and podocin mRNA expression in Gq-KO mice compared to Gq mice. We next measured nephrin protein levels in glomerular preparations by immunoblotting. As shown in Figs 7C and 7D, the pattern of nephrin expression was similar to the quantitative RT-PCR results, but the changes were not statistically significant. Additional mRNA data is presented for WT1, synaptopodin, podocalyxin, TGF-beta and IL11 in Supplementary S2 Table in S1 Data.

Fig 7. Effect of NPRC KO on podocyte proteins.

Fig 7

(7A & 7B) Expression of nephrin and podocin mRNAs were significantly decreased in glomerular preparations from Gq mice compared to controls and expression of both podocyte proteins were preserved by podocyte specific KO of NPRC. (7C & 7D) The pattern of nephrin protein expression was similar to the mRNA results, but the differences were not statistically significant.

Lastly, we evaluated kidney function in Gq mice and controls by measuring cystatin C levels in serum, which is reported to be more precise for assessing renal function in mice compared to serum creatinine levels [39]. As shown in Fig 8, there was a significant increase in cystatin C levels in Gq mice compared to controls. Podocyte specific KO of NPC reduced cystatin C levels compared to Gq mice, but the decrease was not statistically significant.

Fig 8. Effect of NPRC KO on renal function.

Fig 8

Cystatin C levels were significantly increased in serum of Gq mice compared to controls. KO of NPRC decreased cystatin C levels in Gq mice but the difference was not statistically significant.

Discussion

In this study, we found that podocyte specific KO of NPRC had podocyte protective effects in cultured podocytes and ameliorated kidney disease in a mouse model of FSGS. In the kidney, KO of NPRC: 1. Decreasing albuminuria, GS, and TI inflammation, 2. Preserving expression of the podocyte proteins nephrin and podocin, and 3. Inhibiting expression of the myofibroblast marker alpha-SMA and the ECM proteins fibronectin and Col4 alpha-1. The beneficial effects of NPRC KO were accomplished without a significant difference in systemic blood pressure between Gq mice and Gq-KO mice. These data suggest that podocyte specific KO of NPRC had multiple beneficial effects in an FSGS model. Taken together, we posit that inhibiting NP clearance to enhance the effects of endogenous NPs may be a useful therapeutic approach to treat glomerular disease processes.

The biological effects of NPs are modulated by multiple mechanisms in addition to clearance of NPs by NPRC including: 1. The protease neprilysin that cleaves circulating NPs [11], and 2. Intracellular hydrolysis of cGMP by phosphodiesterases (PDEs) [11,40]. All these mechanisms likely play significant roles in regulating the biologic effects of NPs. We previously found that: 1. NPRC and neprilysin are expressed in cultured podocytes [17], in agreement with previous studies [8,14,15,17,27], 2. Cultured podocytes express PDEs important for regulating intracellular levels of cGMP including PDE5 and PDE9 [17], and 3. Pharmacologic blockade of NPRC in cultured podocytes enhanced cGMP generation to a greater extent than pharmacologic inhibition of either PDE5, PDE9 or neprilysin inhibition, using maximally effective doses of each drug [17]. These results agree with in vivo studies that demonstrated pharmacologic blockade of NPRC increased ANP more effectively than neprilysin antagonists [10,17,4144]. These data suggest that targeting NPRC is an effective strategy to reduce clearance of NPs from the circulation by NPRC and enhance NP signaling.

Both ANP and BNP bind to NPRA, stimulate cGMP generation, and have renal protective actions in animal models of kidney disease [15,18,21,22]. Less is known about potential renal protective effects of CNP. Moreover, CNP is expressed by multiple cell types including podocytes (visceral epithelial cells) [12,25] and may have local renal protective actions. In cultured podocytes, we found that KD of NPRC enhanced cGMP generation by low doses of CNP (Fig 1F) and reduced apoptosis induced in the absence of treatment with exogenous NPs (Fig 1G). The podocyte protective action of NPRC KD was partially attenuated by pharmacologic blockade of NPRB in KD cells using the NPRB antagonist P19 (Fig 1G) [29,30]. Interpretation of the P19 experiments is, however, dependent on the specificity of P19 blockade [29]. In the KD cells, binding of P19 to either NPRC or NPRA is unlikely to affect the apoptotic response because: 1. NPRC is absent in the KD cells, and 2. The dosage of P19 used in the studies (250 nM) effectively binds NPRB (Kd 15.4 nM) but has significantly less affinity for NPRA (NPRA Kd 575 nM). In contrast to the KD cells, NPRC is expressed in the control cells, which could affect the apoptotic response by blocking natriuretic clearance and enhancing CNP levels. We posit that blockade of NPRC in the control cells (Fig 1G) was little affected by P19 treatment because P19 has high affinity for both NPRB (Kd ~ 15 nM) and NPRC (0.0134 nM) and, in turn, would effectively inhibit both NP receptors (NPRB and NPRC). In this scenario, the benefits of increasing CNP levels by blockade NPRC would be reduced by pharmacologic blockade of NPRB. Taken together, these data suggest that the podocyte protective actions of NPRC KD are mediated, as least in part, by endogenously produced CNP acting in an autocrine fashion to stimulate NPRB signaling.

We also acknowledge that that NPRC is not only a clearance receptor, but also stimulates intracellular signaling. Following ligand engagement, NPRC stimulates phospholipase C and inhibits both L-type ion channels and cAMP generation [4547]. Theses signaling pathways have been reported to play important roles in cardiovascular disease [46,48], although their role in kidney diseases has not been carefully investigated. As a result, signaling pathways linked to NPRC activation may also contribute to the beneficial effects of NPRC KO.

In addition, we compared the effects of ANP and CNP on both cGMP generation and apoptotic responses. We found that activation of NPRA by ANP was more effective than CNP at inhibiting podocyte apoptosis at equivalent 10 nM concentrations in cultured podocytes (Fig 2D). While we can only speculate about mechanisms, previous studies suggest that ANP and CNP receptors bind their respective ligands with similar, high affinities [11,49] and, while single cell sequencing studies suggest that NPRA expression is more abundant in podocytes compared to NPRB [14,16,50], the levels of NPRA and NPRB were similar in cultured podocytes (Fig 2B) with the exception of the decrease in NPRB levels in the NPRC KD cells (Fig 2B) (discussed below). The culture system does, however, have significant differences from the in vivo situation. CNP is produced by podocytes [12,25] and CNP levels in culture medium were enhanced in NPRC KD cells (Figs 1D, 1C). Chronic exposure to NPs causes homologous desensitization [11,51], which causes a decrease in NPR responsiveness. In this scenario, continuous stimulation of NPRB by CNP may attenuate NPRB signaling. Moreover, prolonged exposure to ligand could theoretically cause receptor downregulation [51]. While downregulation of NPRs is controversial [10,52], recent studies suggest that prolonged treatment with ANP downregulates NPRA [53]. In a similar fashion, chronically enhanced CNP levels might explain the decrease in NPRB levels (Fig 2B) in cultured NPRC KD podocytes (Fig 2B).

To determine if enhancing NP signaling was podocyte protective in vivo, we examined the beneficial effects of NPRC KO in a mouse model of FSGS. We found that podocyte specific KO of NPRC reduced proteinuria, improved renal histology and decreased expression of the myofibroblast marker alpha-SMA and the ECM proteins fibronectin and Col4 alpha-1. The signaling pathways that regulate expression of these fibrotic markers are complex, but NPs inhibit several pathways that promote fibrosis. For example, NPs are potent inhibitors of Rho A [54], which decreases both fibroblast proliferation [11] and differentiation of fibroblasts into myofibroblasts [54]. In addition, cGMP signaling has diverse effects on calcium signaling [9] including: 1. Directly phosphorylating and inhibiting ion channels that increase intracellular calcium levels such as TRPC6 [5558], 2. Sequestering calcium in the sarcoplasmic reticulum [59], and 3. Inhibiting calcium release from intracellular stores [60,61]. As a result, calcium signaling is impaired in multiple receptors implicated in promoting renal fibrosis including angiotensin II and endothelin [9,11,54,62,63]. Lastly, NPs also inhibit the master regulator of fibrosis, TGF-beta, by phosphorylating SMADs on an inhibitory site [20]. Thus, multiple potential mechanisms may contribute to the beneficial effects of NP signaling on renal fibrosis.

As mentioned above, NPRC KO decreased the expression of the ECM protein fibronectin (Figs 6G & 6H), but had only a modest, and non-significant effect on fibronectin mRNA expression (Fig 6B). While fibronectin is produced within the glomerulus, the protein also circulates in blood and is deposited in glomeruli in kidney diseases including FSGS [64] and diabetic kidney disease [65]. The predominant source of circulating fibronectin is the liver [64] and, in animal models, the major source of fibronectin in early stages of FSGS is from the circulation with deposition of glomerular fibronectin occurring later in the disease process [64]. Given the duration of the current animal studies (14 days), the increase in glomerular fibronectin in the present study was likely caused by deposition of fibronectin from the circulation, with lesser amounts produced in the glomeruli. We suspect that the discrepancy in the mRNA expression and protein levels is related to greater deposition of fibronectin from the circulation in damaged glomeruli compared to glomeruli with less severe glomerular injury.

We also found a significant decrease in TI inflammation in Gq-KO mice (Fig 5D). A similar improvement in the accumulation of inflammatory cells was observed by stimulating soluble guanylate cyclase and, in turn, enhancing cGMP generation in a rat model of anti-Thy1-induced glomerulonephritis [66]. Indeed, the same signaling pathways that stimulate myofibroblast differentiation (TGF-beta, Rho GTPases, and calcium) may also promote inflammation by modulating immune cell migration and the severity of immune response [6769]. The level of proteinuria may also contribute to TI inflammation. In this regard, filtered proteins are resorbed by the proximal tubules and processed by lysosomes and endoplasmic reticulum [70]. In the setting of heavy proteinuria, these intracellular pathways are stressed [71], which results in the secretion of cytokines that both attract and activate inflammatory cells [72] and produce profibrotic mediators such as TGF-beta [73]. These data suggest that the anti-inflammatory actions of the NPRC inhibition may play an important role in the beneficial actions of this treatment approach.

The evidence that NPRC clears NPs from the circulation was initially based on: 1.Pharmacologic blockade of NPRC increased ANP plasma levels in rats [74], and 3. KO of NPRC increased the half-life of ANP in the circulation in mice [13]. These data suggested that a major function of NPRC was to negatively regulate the effects of ANP. More recent studies indicate that other NP family members are negatively regulated by the clearance receptor (NPRC). For example, mutations that disrupt the function of genes encoding either CNP or NPRB (CNP receptor) causes dwarfism [7577]. In contrast, disruption of the gene encoding the clearance receptor (NPRC) caused bone overgrowth [13]. These findings indicate: 1. A major function of CNP is binding to NPRB and stimulating bone growth, and 2. The stimulatory effects of CNP on bone growth are augmented by decreased clearance of CNP from the circulation by NPRC. Taken together, these data are consistent with the notion that multiple NP family members are negatively regulated by NPRC.

Lastly. there have been several clinical studies examining the effects of enhancing NP signaling on progression of chronic kidney disease (CKD) using a drug combination that contained both the neprilysin inhibitor sacubitril and the angiotensin receptor blocker (ARB) valsartan [7880]. Two of these manuscripts [78,79] were secondary analyses of studies in heart failure patients that compared sacubitril/valsartan to renin-angiotensin system inhibition alone. Both these secondary analyses found a significant reduction in the rate of decline in the glomerular filtration rate (GFR) in patients receiving the drug combination (sacubitril/valsartan) compared to the control group [78,79]. Interpretation of these studies is, however, complicated by the significant beneficial effects of sacubitril/valsartan on heart failure, which may have improved renal hemodynamics. A third study investigated the effects of sacubitril/valsartan versus the ARB irbesartan in patients with CKD using a randomized, double-blind study design [80]. In this trial, treatment with sacubitril/valsartan caused a significant decrease in systolic and diastolic blood pressure but no clear improvement in GFR or proteinuria [80,81]. Interpretation of these results is complicated because of the short duration of the study, the low number of patients enrolled in the trial and the diverse study population that included a large number of patients without glomerular diseases.

In summary, knockdown of NPRC enhanced the podocyte protective actions of both ANP and CNP in cultured podocytes, and podocyte specific KO of NPRC had beneficial effects on proteinuria, GS, TI inflammation and expression of slit diaphragm and ECM proteins in a mouse model of FSGS. These data suggest that inhibiting NP clearance by NPRC might be a useful therapeutic approach to treat glomerular diseases.

Methods

Primary antibodies used for the studies included: 1. A mouse monoclonal antibody to alpha-smooth muscle actin [82] (clone 1A4, catalog number: A5228, Sigma-Aldrich, St. Louis, MO), 2. A mouse monoclonal antibody to actin [83] (clone C4, catalog number: MA1501, Sigma-Aldrich, St. Louis, MO), 3. A rabbit polyclonal antibody to fibronectin (catalog number: ab2413, Abcam biotechnology, Cambridge, United Kingdom), 4. A goat polyclonal antibody to nephrin [84] (catalog number: AF3159, R&D Systems, Minneapolis, MN), 5. A rabbit monoclonal antibody to WT1 (Wilms tumor 1) (catalog number: ab89901, Abcam Biotechnology, Cambridge, United Kingdom), 6. A mouse monoclonal IgG antibody to the podocyte marker synaptopodin (catalog number 65194, Progen Biotechnik, Heidelberg, Germany), 7. A mouse monoclonal IgG antibody to NPRC (clone OTI4H1, catalog number: TA501044, Origene Technologies, Rockville, MD) and a polyclonal Goat IgG antibody to CNP (AF3127, R&D Systems, Minneapolis, MN). Secondary antibodies used for the studies included: 1. A mouse HRP-linked anti-goat polyclonal antibody (catalog number: sc-2354, Santa Cruz Biotechnology, Dallas, TX), and 2. An anti-mouse HRP-linked polyclonal antibody (catalog #: 7076) and an anti-rabbit HRP-linked polyclonal antibody (catalog #: 7074) from Cell Signaling Technology, Danvers, MA). 3. A Cy3 labeled donkey polyclonal anti-goat IgG antibody (Abcam Biotechnology, Cambridge, United Kingdom), and 4. A FITC labeled polyclonal donkey anti-mouse IgG antibody (Abcam Biotechnology, Cambridge, United Kingdom). Additional materials included: 1. Albuwell and Creatinine Companion kits (catalog No. 1011 and 1012, respectively, Ethos Biosciences, Logan Township, NJ), 2. Rat CNP (Item No. 24276) and cGMP ELISA kits (Item No.581021) from Cayman Chemical, Ann Arbor, MI, 3. Annexin V apoptosis kits, (catalog No. 559763, BD Pharmingen, Franklin Lakes, NJ), and 4. Recombinant mouse IFN-gamma (catalog No. 485-MI-100, R&D Systems, Minneapolis, MN).

BP measurements

Systolic BP was measured using a computerized tail-cuff system (Hatteras Instruments, Cary, NC, USA) in conscious mice as previously described [85]. To reduce variability in the results, mice were acclimated to the experimental conditions for a week prior to the BP measurements. This technique has previously been shown to correlate closely with intra-arterial measurements [86].

Animal subjects

The following mouse models were used for the studies: 1. GqQ > L mice were created in our laboratory as described [23], 2. (NPRC-flox/flox mice were obtained from Taconic Biosciences, No. 13105) and bred onto the FVB background for over 6 generations, 3. A doxycycline inducible Cre recombinase mouse line was obtained from Dr. Jeffery Kopp on the FVB background [87] (NOTE: This mouse are now available from Jackson Labs (tetO-Cre, No. 008205). The tdTomato mouse [31] was originally obtained from Jackson Labs (Stock No. 007576).

Animal experiments

Animal experiments were performed using a transgenic (TG) mouse model of FSGS developed in our laboratory (Gq mice) [23]. The model was created by generating mice with 2 transgenes: 1. The reverse tetracycline transactivator (rtTA) under the control of the podocyte specific podocin promoter (NPHS2-rtTA, Jackson Labs, catalog No. 008202), and 2. GqQ > L under the control of the doxycycline inducible tet operator sequence (tetO) and a minimal cytomegalovirus (CMV) promoter (tetO-GqQ > L). To create a podocyte specific NPRC KO in this TG model, we bred GqQ > L mice with: 1. Animals expressing two NPRC floxed alleles (NPRC-f/f mice, Taconic Biosciences, No. 13105), and 2. A doxycycline inducible Cre recombinase line from Jackson Labs (tetO-Cre, No. 008205). This breeding strategy generated TG mice on the FVB/NJ background expressing 3 transgenes (NPHS2-rtTA, tetO-Cre, tetO-GqQ > L). In “triple” TG mice, treatment doxycycline in 2% sucrose water induces podocyte specific GqQ > L expression and NPRC-KO on demand. All animals were breed onto the FVB/NC background for over 6 generations. A schematic of the breeding strategy is shown in S7 Fig.

For the experiments, age and sex-matched male and female mice were used for the studies because the phenotype is similar in both sexes [17,23]. Mice were studied at 3-4 months of age and all mice were born within a 5-week period. Baseline twenty-four-hour urine collections were collected using metabolic cages specifically designed for collection of mouse urine (Hatteras Instruments, Cary, NC). After collecting baseline urines, mice were treated with doxycycline for 1 week and then nephrosis was induced by a single injection of PAN (500 mg/kg) by intraperitoneal (IP) injection. Doxycycline was continued for an additional 2 weeks. Systolic blood pressure (SBP) was measured as described [85] during the second week after the PAN injection, and following acclimation to the experimental procedure during the prior week. Repeat 24-hour urine collections were obtained at day 10 and day 14 following the PAN injection. Mice were euthanized after the last urine collection by injecting 250 mg/kg pentobarbital, IP, followed by bilateral thoracotomy. Blood was immediately obtained by cardiac puncture after euthanasia and then kidneys were removed, weighed and kidney tissue was saved for additional studies as described below. The experiments conformed to the Guide for the Care and Use of Laboratory Animals and were approved by the Duke and Durham VA Medical Centers’ Institutional Animal Care and Use Committees. A schematic of the experimental protocol is provided in supplementary S3 Fig.

Podocyte specific NPRC KO in td-tomato mice

To assess NPRC KO, we used a reporter mouse available from Jackson labs (Stock Nos. 007576) [85]. The reporter mice express a two-color fluorescent Cre reporter allele containing a cell membrane-localized red fluorescence protein (tdTomato) with widespread expression in all tissues and cell types prior to Cre-mediated recombination (JAX: Stock Nos. 007576). Induction of Cre recombinase in podocytes induces a cell membrane-localized green fluorescence protein (EGFP). For the studies, we crossed the reporter mice with mice expressing NPHS2-rtTA and tetO-Cre to create mice expressing 3 transgenes (tdTomato, NPHS2-rtTA, and tetO-Cre). We then assessed the effectiveness of podocyte specific NPRC-KO by studying mice treated with either doxycycline or vehicle as described above.

Histopathology

After fixation in formalin, light microscopic sections were stained with hematoxylin and eosin (H&E) and Masson trichrome by the Duke Research Animal Pathology Service. The slides were evaluated by an experience medical nephropathologist (A.F.B.) blinded to genotype. Each tissue section had more than twenty glomeruli for evaluation. Tubules were examined for tubule dilation and casts, and tubulointerstitial areas were examined for inflammation with or without interstitial fibrosis. There is little tubulointerstitial fibrosis observed in this model over a 14-day period, and this score was based on the severity of inflammation as described below. Abnormalities were graded using a semi-quantitative scale of 1-4 (1-normal, 2-mild, 3-moderate, 4-severe) as previously described [23,32] based on the following criteria:

Glomerulosclerosis.

  1. Normal (baseline): None

  2. Mild: < 10% of glomeruli

  3. Moderate: 10-25% of glomeruli

  4. Severe: > 25% of glomeruli

Tubule injury.

  1. Normal (baseline): None to minimal tubular dilation and casts without tubular degeneration or regeneration

  2. Mild: Tubular degeneration and regeneration with/without tubular dilation and casts, involving < 10% of cortex

  3. Moderate: Tubular degeneration and regeneration with/without tubular dilation and casts, involving 10–25% of cortex

  4. Severe: Tubular degeneration and regeneration with/without tubular dilation and casts, involving > 25% of cortex

Tubulointerstitial inflammation.

  1. Normal (baseline): One focus of inflammation (up to 15 mononuclear cells) with or without interstitial fibrosis

  2. Mild: Two foci of 15 + mononuclear cells or 1 focus of 30 + mononuclear cells involving up to 5% of area of cortical parenchyma with or without interstitial fibrosis

  3. Moderate: Inflammation involving > 5–25% of the cortical parenchyma with or without interstitial fibrosis

  4. Severe: Inflammation involving > 25% of the cortical parenchyma with or without interstitial fibrosis

Immunofluorescence studies

Expression of human CNP in kidney sections was detected by immunofluorescence imaging using a goat polyclonal IgG antibody to human CNP (AF3127, R&D Systems), and a mouse monoclonal IgG antibody to the podocyte marker synaptopodin (Progen Biotechnik). Normal human kidney frozen sections were obtained from Zyagen (San Diego, CA) and were fixed in 2% paraformaldehyde for 5 minutes, air-dried, treated with 0.1% Triton-X in Dulbecco’s phosphate buffered saline (D-PBS) for 5 minutes and then blocked for 1 hour in D-PBS with 5% bovine serum albumin (BSA). The CNP and synaptopodin primary antibodies were then added at a 1:200 dilution and 1:50, respectively in D-PBS with 5% BSA. After an overnight incubation, slides were washed 3 times in D-PBS and then incubated for 1 hour with a Cy3 labeled donkey polyclonal anti-goat antibody (Abcam Biotechnology), and a FITC labeled polyclonal donkey anti-mouse antibody (Abcam Biotechnology), both at a dilution of 1:1000 in D-PBS with 5% BSA. Sides were then washed 3 times in D-PBS, and coverslips were affixed using with Vectashield (Vector Labs, Newark, CA) mounting medium with DAPI (4’,6-diamidino-2-phenylindole). Slide were examined by dual fluorescence microscopy using a Nikon Eclipse TB-2000 microscope.

Cell culture studies

The immortalized mouse podocyte cell line was a gift of Dr. Paul E. Klotman, MD [88]. The cells were derived from animals bred with the H-2Kb-tsA58 Immortomice (Charles River Laboratories, Wilmington, MA). Podocytes were selected for expression of the podocyte markers WT-1, synaptopodin and podocalyxin. To permit immortalized growth, cells were grown at 33°C in medium supplemented with 10 units/ml gamma-interferon to induce the H-2Kb promoter driving synthesis of the temperature sensitive (tsA58) SV-40 T antigen (permissive conditions). For differentiation, cells were grown at 37°C in medium lacking gamma-interferon, resulting in degradation of the T antigen (non-permissive conditions). Our lab further characterized differentiated cells by RT-PCR, and they also express low levels of the podocyte proteins nephrin and podocin. For the experiments, the immortalized mouse podocyte cell line was maintained in culture and was plated in either 6- or 12- well tissue culture clusters (Corning-Costar, Corning, NY) and differentiated for ~ 7 days prior to study as previously described.

To knockdown (KD) NPRC, we used four shRNA constructs (catalog numbers TG514036A-D; Origene Technologies, Rockville, MD) and a scrambled control (TR30014). The constructs were introduced into our mouse podocyte cell line using Lipofectamine 2000 according to the manufacturer’s recommendations (catalog number 11668027, ThermoFisher Scientific, Waltham, MA), and then selected in 4µg/ml puromycin (catalog number: sc374043, Santa Cruz Biotechnology, Dallas, TX),

Measurement of CNP in cell culture medium

For the study, KD cells and controls were plated in 12-well tissue culture clusters and differentiated for 7 days. After differentiation, cells were washed and placed in serum free medium warmed to 37°C, in the presence or absence of the neprilysin inhibitor LBQ657 (10 µ M final) [28]. After incubation at 37°C for 30 minutes, the medium was aspirated, aliquoted into microfuge tubes, placed on ice and immediately stored at -80°C. CNP in culture medium was measured using an ELISA kit (RK02693) from ABcclonal technologies (Woburn, MA) and data was expressed as pmol/mg protein.

Measurement cGMP in cell culture medium

For the cGMP studies, differentiated podocytes were made quiescent by incubation in serum free medium overnight. Cells were then treated with vehicle or the following concentrations of CNP (0.1, 1.0 and 10 nM). After 30 minutes, the supernatant was aspirated, and the tissue culture clusters placed on ice. Hydrochloric acid (0.1 N) was then added to the wells (100µl per 12 wells and 300µl per 6 well tissue culture cluster). A cell scrapper was used to remove the cells from the cell culture wells, and the mixture dissociated by pipetting up and down until homogeneous. The mixture was then centrifuged at 1000 x g for 10 minutes and the supernatant frozen at -80°C. Measurement of cGMP in the supernatants was performed using an ELISA kit from Cayman Chemicals (Ann Arbor, MI) according to the directions of the manufacturer and data was expressed as pmol/mg protein.

Apoptosis studies

For the apoptosis studies, differentiated mouse podocytes were changed to medium with 1% fetal bovine serum (FBS) or serum free medium. After 2 days of serum deprivation, cells were harvested, and apoptotic cells were identified by annexin V staining using kits from BD Pharmingen (San Diego, CA) according to the directions of the manufacturer. Quantitation of the apoptotic cells was performed by flow cytometric analysis at the Duke Comprehensive Cancer facility. For the annexin V studies, apoptotic podocytes were differentiated from necrotic cells by staining with 7-amino-actinomycin D. For evaluation of these data, basal levels of apoptosis (cells treated with 0.1% FBS and vehicle) were subtracted from the results, and data expressed as the percent apoptosis above basal.

For the pharmacologic studies, P19 (250 nM) or vehicle was added to the culture medium. In addition, the culture medium with P19 or vehicle was replenished twice per day to minimize degradation of the P19 during the study.

Immunoblotting of enriched glomerular preparations

Immunoblotting was performed as previously described [85] using the antibodies listed in Materials. Briefly, enriched glomerular pellets were solubilized by sonication in NP-40 lysis buffer (50 mM Tris-HCl, 150 mM sodium chloride, 2mM ethylenediaminetetraacetic acid, 1% NP-40 and protease inhibitors from Sigma-Aldrich), and frozen at -80°C until the time of study. Proteins were separated using the XCell SureLock Bis-Tris Mini-Cell Electrophoresis System (Thermo Fisher Scientific, Waltham, MA) and transferred to PVDF (polyvinylidene fluoride) membranes according to the directions of the manufacturer. Immunoblots were then blocked in 20 mM Tris-HCl, 137 mM NaCl, pH 7.6 (TBS) with 0.2% Tween 20 (T-TBS) and 2% bovine serum albumin. The primary antibody was added at a 1:1000 dilution in blocking buffer and incubated overnight. After washing, the HRP (horseradish peroxidase) linked secondary antibodies were added at a 1:2000 concentration in blocking solution and incubated for 1 hour prior to washing. Protein detection was performed using enhanced chemiluminescence (ECL) (Thermo Scientific, Waltham, MA) according to the directions of the manufacturer. Imaging of Western blots was performed using a Bio-Rad ChemiDoc-MP imaging system. To assess protein-loading immunoblots were stripped using Restore Western Blot Stripping Buffer (Thermo Scientific, Waltham, MA) according to the directions of the manufacturer and immunoblotting was performed using mouse monoclonal antibodies to b-actin (0.5µg/ml) in blocking solution and an HRP-linked anti-mouse secondary antibody (1:2000). Densitometry was performed using ScanAnalysis 2.5 software (Biosoft). For the analyses, the densitometric data for the protein signal were divided by the matched signal for b-actin to compare separate immunoblots, densitometric data were normalized to WT controls.

Expression of glomerular mRNAs

Reverse transcription (RT) followed by a quantitative polymerase chain reaction (Q-RT-PCR) was performed using an iCycler TM (Bio- Rad Laboratories, Inc., Hercules, CA). For the studies, total cellular RNA was prepared using glomerular preparations and the Trizol reagent according to the manufacturer’s directions (Life Technologies Inc., Carlsbad, CA). The RNA was treated with RNAase free DNAase (Qiagen) and then reverse-transcribed with Superscript reverse transcriptase (Invitrogen) and oligo (dT) primers. Real-time quantitative PCR was performed using an iCycler Q-PCR machine and the universal SYBR Green PCR Master Mix Kit (Perkin-Elmer, Waltham, MA). The amplification signals were normalized to the endogenous GAPDH mRNA level. The primer sequences used for Q-RT-PCR were as follows: CNP forward: 5’- AAT ACA AAG GCG GCA ACA AG – 3’ & reverse, 5’ – TAA CAT CCC AGA CCG CTC AT – 3’. NPRC forward, 5’ – AGC TGG CTA CAG CAA GAA GG – 3’ & reverse, 5’ – CGG CGA TAC CTT CAA ATG TC – 3’; GAPDH, 5’_- GTGAAGGTCGGTGTG AACGGATTTG – 3’ & antisense 5’ -ACATTGGGGGTAGGAACACGGAAGG - 3’; Fibronectin forward 5’ – CGA GGT GAC AGA GAC CAC AA – 3’ & reverse 5’ – CTG GAG TCA AGC CAG ACA CA – 3’; collagen type 1, alpha-1 (COL1A1) forward 5’ – ATC TCC TGG TGC TGA TGG AC – 3’ & reverse 5’ – ACC TTG TTT GCC AGG TTC AC – 3’; alpha-smooth muscle actin (SMA) forward 5’ – GAG GCA CCA CTG ACC CCT AA – 3’ & reverse 5’ –CAT CTC CAG AGT CCA GCA CA – 3’. Collagen 4, alpha 1 (COL4A1) forward 5’ – TATCTCTGGGGACAACATCCG – 3’ & reverse 5’ – CATCTCGCTTCTCTCTATGGTG – 3’, Nephrin forward 5’ –– AGCTACCCTGCATAGCCAGA – 3’ & reverse 5’ – – CCCAAGCTATGGACACTGGT – 3’, Collagen 4, alpha 4 (COL4A4) forward 5’ – CTCCTGGTTCTCCACAGTCAG – 3’ & reverse 5’ – AAGGGCAGAGTGCTAAACACA – 3’, Podocin forward 5’ – GTGTCCAAAGCCATCCAGTT – 3’ & reverse 5’ – GCAATGCTCTTCCTTTCCAG – 3’.

Statistical analysis

Data are presented as the mean ±  standard error of the mean (SEM) and statistical analyses were performed using the Prism 9 computer program (GraphPad Software, Inc.) assuming a normal distribution of the data. For comparison of two groups of continuous variables, data was analyzed by a t-test. For comparison of more than 2 groups of continuous variables, data was analyzed by either a one-way or two-way analysis of variance (ANOVA) as specified by the Prism program, followed by Sidak’s multiple comparisons post-test. For non-continuous variables (histopathology), data was analyzed using a Fishers Exact test using the number mice with the specified histologic abnormality. Graphs of the histopathologic findings were, however, presented as the percentage of mice with the specified abnormality to permit a more effective comparison of the differences between the experimental groups in studies with an imbalance in the number of mice in each group. All statistics were performed using two-sided tests.

Supporting information

S1 Data

(PDF)

pone.0319424.s001.pdf (1.4MB, pdf)

Data Availability

All relevant data are within the manuscript and its Supporting Information files.

Funding Statement

1. R21 TR004257-01 from the National Institutes of Health. 2. BX002984 from the Veterans Administration Merit Review Program. 3. W81XWH-19-1-0314 from the United States Department of Defense. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Rosenberg AZ, Kopp JB. Focal segmental glomerulosclerosis. Clin J Am Soc Nephrol. 2017;12(3):502–17. doi: 10.2215/CJN.05960616 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.D’Agati VD, Kaskel FJ, Falk RJ. Focal segmental glomerulosclerosis. N Engl J Med. 2011;365(25):2398–411. doi: 10.1056/NEJMra1106556 [DOI] [PubMed] [Google Scholar]
  • 3.Korbet SM. Treatment of primary FSGS in adults. J Am Soc Nephrol. 2012;23(11):1769–76. doi: 10.1681/ASN.2012040389 [DOI] [PubMed] [Google Scholar]
  • 4.The EMPA-KIDNEY Collaborative Group, Herrington WG, Staplin N, Wanner C, Green JB, Hauske SJ, et al. Empagliflozin in patients with chronic kidney disease. N Engl J Med. 2023;388(2):117–27. doi: 10.1056/NEJMoa2204233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Heerspink HJL, Stefánsson BV, Correa-Rotter R, Chertow GM, Greene T, Hou F-F, et al. Dapagliflozin in patients with chronic kidney disease. N Engl J Med. 2020;383(15):1436–46. doi: 10.1056/NEJMoa2024816 [DOI] [PubMed] [Google Scholar]
  • 6.Nuffield Department of Population Health Renal Studies Group, SGLT2 inhibitor Meta-Analysis Cardio-Renal Trialists’ Consortium. Impact of diabetes on the effects of sodium glucose co-transporter-2 inhibitors on kidney outcomes: collaborative meta-analysis of large placebo-controlled trials. Lancet. 2022;400(10365):1788–801. doi: 10.1016/S0140-6736(22)02074-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Wiggins RC. The spectrum of podocytopathies: a unifying view of glomerular diseases. Kidney Int. 2007;71(12):1205–14. doi: 10.1038/sj.ki.5002222 [DOI] [PubMed] [Google Scholar]
  • 8.Wolf G, Chen S, Ziyadeh FN. From the periphery of the glomerular capillary wall toward the center of disease: podocyte injury comes of age in diabetic nephropathy. Diabetes. 2005;54(6):1626–34. doi: 10.2337/diabetes.54.6.1626 [DOI] [PubMed] [Google Scholar]
  • 9.Francis SH, Busch JL, Corbin JD, Sibley D. cGMP-dependent protein kinases and cGMP phosphodiesterases in nitric oxide and cGMP action. Pharmacol Rev. 2010;62(3):525–63. doi: 10.1124/pr.110.002907 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Potter LR. Natriuretic peptide metabolism, clearance and degradation. FEBS J. 2011;278(11):1808–17. doi: 10.1111/j.1742-4658.2011.08082.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Potter LR, Abbey-Hosch S, Dickey DM. Natriuretic peptides, their receptors, and cyclic guanosine monophosphate-dependent signaling functions. Endocr Rev. 2006;27(1):47–72. doi: 10.1210/er.2005-0014 [DOI] [PubMed] [Google Scholar]
  • 12.Lewko B, Endlich N, Kriz W, Stepinski J, Endlich K. C-type natriuretic peptide as a podocyte hormone and modulation of its cGMP production by glucose and mechanical stress. Kidney Int. 2004;66(3):1001–8. doi: 10.1111/j.1523-1755.2004.00848.x [DOI] [PubMed] [Google Scholar]
  • 13.Matsukawa N, Grzesik WJ, Takahashi N, Pandey KN, Pang S, Yamauchi M, et al. The natriuretic peptide clearance receptor locally modulates the physiological effects of the natriuretic peptide system. Proc Natl Acad Sci U S A. 1999;96(13):7403–8. doi: 10.1073/pnas.96.13.7403 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Park J, Shrestha R, Qiu C, Kondo A, Huang S, Werth M, et al. Single-cell transcriptomics of the mouse kidney reveals potential cellular targets of kidney disease. Science. 2018;360(6390):758–63. doi: 10.1126/science.aar2131 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Staffel J, Valletta D, Federlein A, Ehm K, Volkmann R, Füchsl AM, et al. Natriuretic peptide receptor guanylyl cyclase-a in podocytes is renoprotective but dispensable for physiologic renal function. J Am Soc Nephrol. 2017;28(1):260–77. doi: 10.1681/ASN.2015070731 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wilson PC, Wu H, Kirita Y, Uchimura K, Ledru N, Rennke HG, et al. The single-cell transcriptomic landscape of early human diabetic nephropathy. Proc Natl Acad Sci U S A. 2019;116(39):19619–25. doi: 10.1073/pnas.1908706116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wang L, Tang Y, Buckley AF, Spurney RF. Blockade of the natriuretic peptide clearance receptor attenuates proteinuria in a mouse model of focal segmental glomerulosclerosis. Physiol Rep. 2021;9(21):e15095. doi: 10.14814/phy2.15095 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Makino H, Mukoyama M, Mori K, Suganami T, Kasahara M, Yahata K, et al. Transgenic overexpression of brain natriuretic peptide prevents the progression of diabetic nephropathy in mice. Diabetologia. 2006;49(10):2514–24. doi: 10.1007/s00125-006-0352-y [DOI] [PubMed] [Google Scholar]
  • 19.Ogawa Y, Mukoyama M, Yokoi H, Kasahara M, Mori K, Kato Y, et al. Natriuretic peptide receptor guanylyl cyclase-A protects podocytes from aldosterone-induced glomerular injury. J Am Soc Nephrol. 2012;23(7):1198–209. doi: 10.1681/ASN.2011100985 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Shen K, Johnson DW, Gobe GC. The role of cGMP and its signaling pathways in kidney disease. Am J Physiol Renal Physiol. 2016;311(4):F671–81. doi: 10.1152/ajprenal.00042.2016 [DOI] [PubMed] [Google Scholar]
  • 21.Suganami T, Mukoyama M, Sugawara A, Mori K, Nagae T, Kasahara M, et al. Overexpression of brain natriuretic peptide in mice ameliorates immune-mediated renal injury. J Am Soc Nephrol. 2001;12(12):2652–63. doi: 10.1681/ASN.V12122652 [DOI] [PubMed] [Google Scholar]
  • 22.Kato Y, Mori K, Kasahara M, Osaki K, Ishii A, Mori KP, et al. Natriuretic peptide receptor guanylyl cyclase-A pathway counteracts glomerular injury evoked by aldosterone through p38 mitogen-activated protein kinase inhibition. Sci Rep. 2017;7:46624. doi: 10.1038/srep46624 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Wang L, Jirka G, Rosenberg PB, Buckley AF, Gomez JA, Fields TA, et al. Gq signaling causes glomerular injury by activating TRPC6. J Clin Invest. 2015;125(5):1913–26. doi: 10.1172/JCI76767 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lumsden NG, Khambata RS, Hobbs AJ. C-type natriuretic peptide (CNP): cardiovascular roles and potential as a therapeutic target. Curr Pharm Des. 2010;16(37):4080–8. doi: 10.2174/138161210794519237 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Cataliotti A, Giordano M, De Pascale E, Giordano G, Castellino P, Jougasaki M, et al. CNP production in the kidney and effects of protein intake restriction in nephrotic syndrome. Am J Physiol Renal Physiol. 2002;283(3):F464–72. doi: 10.1152/ajprenal.00372.2001 [DOI] [PubMed] [Google Scholar]
  • 26.Kubiak-Wlekły A, Perkowska-Ptasińska A, Olejniczak P, Rochowiak A, Kaczmarek E, Durlik M, et al. The Comparison of the Podocyte Expression of Synaptopodin, CR1 and Neprilysin in Human Glomerulonephritis: Could the Expression of CR1 be Clinically Relevant?. Int J Biomed Sci. 2009;5(1):28–36. doi: 10.59566/ijbs.2009.5028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Debiec H, Guigonis V, Mougenot B, Decobert F, Haymann J-P, Bensman A, et al. Antenatal membranous glomerulonephritis due to anti-neutral endopeptidase antibodies. N Engl J Med. 2002;346(26):2053–60. doi: 10.1056/NEJMoa012895 [DOI] [PubMed] [Google Scholar]
  • 28.Campbell DJ. Long-term neprilysin inhibition—implications for ARNIs. Nat Rev Cardiol. 2017;14(3):171–86. doi: 10.1038/nrcardio.2016.200 [DOI] [PubMed] [Google Scholar]
  • 29.Deschênes J, Duperé C, McNicoll N, L’Heureux N, Auger F, Fournier A, et al. Development of a selective peptide antagonist for the human natriuretic peptide receptor-B. Peptides. 2005;26(3):517–24. doi: 10.1016/j.peptides.2004.10.017 [DOI] [PubMed] [Google Scholar]
  • 30.Buttgereit J, Shanks J, Li D, Hao G, Athwal A, Langenickel TH, et al. C-type natriuretic peptide and natriuretic peptide receptor B signalling inhibits cardiac sympathetic neurotransmission and autonomic function. Cardiovasc Res. 2016;112(3):637–44. doi: 10.1093/cvr/cvw184 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Muzumdar MD, Tasic B, Miyamichi K, Li L, Luo L. A global double-fluorescent Cre reporter mouse. Genesis. 2007;45(9):593–605. doi: 10.1002/dvg.20335 [DOI] [PubMed] [Google Scholar]
  • 32.Wang L, Chang J-H, Buckley AF, Spurney RF. Knockout of TRPC6 promotes insulin resistance and exacerbates glomerular injury in Akita mice. Kidney Int. 2019;95(2):321–32. doi: 10.1016/j.kint.2018.09.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Cai YI, Sich M, Beziau A, Kleppel MM, Gubler MC. Collagen distribution in focal and segmental glomerulosclerosis: an immunofluorescence and ultrastructural immunogold study. J Pathol. 1996;179(2):188–96. doi: [DOI] [PubMed] [Google Scholar]
  • 34.Huang A, Guo G, Yu Y, Yao L. The roles of collagen in chronic kidney disease and vascular calcification. J Mol Med (Berl). 2021;99(1):75–92. doi: 10.1007/s00109-020-02014-6 [DOI] [PubMed] [Google Scholar]
  • 35.Lee HS. Oxidized LDL, glomerular mesangial cells and collagen. Diabetes Res Clin Pract. 1999;45(2–3):117–22. doi: 10.1016/s0168-8227(99)00040-6 [DOI] [PubMed] [Google Scholar]
  • 36.Yoshioka K, Tohda M, Takemura T, Akano N, Matsubara K, Ooshima A, et al. Distribution of type I collagen in human kidney diseases in comparison with type III collagen. J Pathol. 1990;162(2):141–8. doi: 10.1002/path.1711620207 [DOI] [PubMed] [Google Scholar]
  • 37.Ebihara I, Suzuki S, Nakamura T, Fukui M, Yaguchi Y, Tomino Y, et al. Extracellular matrix component mRNA expression in glomeruli in experimental focal glomerulosclerosis. J Am Soc Nephrol. 1993;3(7):1387–97. doi: 10.1681/ASN.V371387 [DOI] [PubMed] [Google Scholar]
  • 38.Genovese F, Manresa AA, Leeming DJ, Karsdal MA, Boor P. The extracellular matrix in the kidney: a source of novel non-invasive biomarkers of kidney fibrosis?. Fibrogenesis Tissue Repair. 2014;7(1):4. doi: 10.1186/1755-1536-7-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Song S, Meyer M, Türk TR, Wilde B, Feldkamp T, Assert R, et al. Serum cystatin C in mouse models: a reliable and precise marker for renal function and superior to serum creatinine. Nephrol Dial Transplant. 2009;24(4):1157–61. doi: 10.1093/ndt/gfn626 [DOI] [PubMed] [Google Scholar]
  • 40.Rotella DP. Phosphodiesterase 5 inhibitors: current status and potential applications. Nat Rev Drug Discov. 2002;1(9):674–82. doi: 10.1038/nrd893 [DOI] [PubMed] [Google Scholar]
  • 41.Charles CJ, Espiner EA, Nicholls MG, Richards AM, Yandle TG, Protter A, et al. Clearance receptors and endopeptidase 24.11: equal role in natriuretic peptide metabolism in conscious sheep. Am J Physiol. 1996;271(2 Pt 2):R373-80. doi: 10.1152/ajpregu.1996.271.2.R373 [DOI] [PubMed] [Google Scholar]
  • 42.Hashimoto Y, Nakao K, Hama N, Imura H, Mori S, Yamaguchi M, et al. Clearance mechanisms of atrial and brain natriuretic peptides in rats. Pharm Res. 1994;11(1):60–4. doi: 10.1023/a:1018941626731 [DOI] [PubMed] [Google Scholar]
  • 43.Kukkonen P, Vuolteenaho O, Ruskoaho H. Basal and volume expansion-stimulated plasma atrial natriuretic peptide concentrations and hemodynamics in conscious rats: effects of SCH 39.370, an endopeptidase inhibitor, and C-ANF-(4-23), a clearance receptor ligand. Endocrinology. 1992;130(2):755–65. doi: 10.1210/endo.130.2.1531129 [DOI] [PubMed] [Google Scholar]
  • 44.Okolicany J, McEnroe GA, Koh GY, Lewicki JA, Maack T. Clearance receptor and neutral endopeptidase-mediated metabolism of atrial natriuretic factor. Am J Physiol. 1992;263(3 Pt 2):F546-53. doi: 10.1152/ajprenal.1992.263.3.F546 [DOI] [PubMed] [Google Scholar]
  • 45.El Andalousi J, Li Y, Anand-Srivastava MB. Natriuretic peptide receptor-C agonist attenuates the expression of cell cycle proteins and proliferation of vascular smooth muscle cells from spontaneously hypertensive rats: role of Gi proteins and MAPkinase/PI3kinase signaling. PLoS One. 2013;8(10):e76183. doi: 10.1371/journal.pone.0076183 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Li Y, Sarkar O, Brochu M, Anand-Srivastava MB. Natriuretic peptide receptor-C attenuates hypertension in spontaneously hypertensive rats: role of nitroxidative stress and Gi proteins. Hypertension. 2014;63(4):846–55. doi: 10.1161/HYPERTENSIONAHA.113.01772 [DOI] [PubMed] [Google Scholar]
  • 47.Murthy KS, Teng B, Jin J, Makhlouf GM. G protein-dependent activation of smooth muscle eNOS via natriuretic peptide clearance receptor. Am J Physiol. 1998;275(6):C1409–16. doi: 10.1152/ajpcell.1998.275.6.C1409 [DOI] [PubMed] [Google Scholar]
  • 48.Khambata RS, Panayiotou CM, Hobbs AJ. Natriuretic peptide receptor-3 underpins the disparate regulation of endothelial and vascular smooth muscle cell proliferation by C-type natriuretic peptide. Br J Pharmacol. 2011;164(2b):584–97. doi: 10.1111/j.1476-5381.2011.01400.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Potter LR. Guanylyl cyclase structure, function and regulation. Cell Signal. 2011;23(12):1921–6. doi: 10.1016/j.cellsig.2011.09.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wu H, Kirita Y, Donnelly EL, Humphreys BD. Advantages of single-nucleus over single-cell rna sequencing of adult kidney: rare cell types and novel cell states revealed in fibrosis. J Am Soc Nephrol. 2019;30(1):23–32. doi: 10.1681/ASN.2018090912 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Potter LR, Hunter T. Guanylyl cyclase-linked natriuretic peptide receptors: structure and regulation. J Biol Chem. 2001;276(9):6057–60. doi: 10.1074/jbc.R000033200 [DOI] [PubMed] [Google Scholar]
  • 52.Fan D, Bryan PM, Antos LK, Potthast RJ, Potter LR. Down-regulation does not mediate natriuretic peptide-dependent desensitization of natriuretic peptide receptor (NPR)-A or NPR-B: guanylyl cyclase-linked natriuretic peptide receptors do not internalize. Mol Pharmacol. 2005;67(1):174–83. doi: 10.1124/mol.104.002436 [DOI] [PubMed] [Google Scholar]
  • 53.Khurana ML, Mani I, Kumar P, Ramasamy C, Pandey KN. Ligand-dependent downregulation of guanylyl cyclase/natriuretic peptide receptor-a: role of miR-128 and miR-195. Int J Mol Sci. 2022;23(21):13381. doi: 10.3390/ijms232113381 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Tsou P-S, Haak AJ, Khanna D, Neubig RR. Cellular mechanisms of tissue fibrosis. 8. Current and future drug targets in fibrosis: focus on Rho GTPase-regulated gene transcription. Am J Physiol Cell Physiol. 2014;307(1):C2-13. doi: 10.1152/ajpcell.00060.2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Takahashi S, Lin H, Geshi N, Mori Y, Kawarabayashi Y, Takami N, et al. Nitric oxide-cGMP-protein kinase G pathway negatively regulates vascular transient receptor potential channel TRPC6. J Physiol. 2008;586(17):4209–23. doi: 10.1113/jphysiol.2008.156083 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Kinoshita H, Kuwahara K, Nishida M, Jian Z, Rong X, Kiyonaka S, et al. Inhibition of TRPC6 channel activity contributes to the antihypertrophic effects of natriuretic peptides-guanylyl cyclase-A signaling in the heart. Circ Res. 2010;106(12):1849–60. doi: 10.1161/CIRCRESAHA.109.208314 [DOI] [PubMed] [Google Scholar]
  • 57.Nishida M, Watanabe K, Sato Y, Nakaya M, Kitajima N, Ide T, et al. Phosphorylation of TRPC6 channels at Thr69 is required for anti-hypertrophic effects of phosphodiesterase 5 inhibition. J Biol Chem. 2010;285(17):13244–53. doi: 10.1074/jbc.M109.074104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Hall G, Rowell J, Farinelli F, Gbadegesin RA, Lavin P, Wu G, et al. Phosphodiesterase 5 inhibition ameliorates angiontensin II-induced podocyte dysmotility via the protein kinase G-mediated downregulation of TRPC6 activity. Am J Physiol Renal Physiol. 2014;306(12):F1442–50. doi: 10.1152/ajprenal.00212.2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Cornwell TL, Pryzwansky KB, Wyatt TA, Lincoln TM. Regulation of sarcoplasmic reticulum protein phosphorylation by localized cyclic GMP-dependent protein kinase in vascular smooth muscle cells. Mol Pharmacol. 1991;40(6):923–31. [PubMed] [Google Scholar]
  • 60.Komalavilas P, Lincoln TM. Phosphorylation of the inositol 1,4,5-trisphosphate receptor. Cyclic GMP-dependent protein kinase mediates cAMP and cGMP dependent phosphorylation in the intact rat aorta. J Biol Chem. 1996;271(36):21933–8. doi: 10.1074/jbc.271.36.21933 [DOI] [PubMed] [Google Scholar]
  • 61.Schlossmann J, Ammendola A, Ashman K, Zong X, Huber A, Neubauer G, et al. Regulation of intracellular calcium by a signalling complex of IRAG, IP3 receptor and cGMP kinase Ibeta. Nature. 2000;404(6774):197–201. doi: 10.1038/35004606 [DOI] [PubMed] [Google Scholar]
  • 62.Horinouchi T, Terada K, Higashi T, Miwa S. Endothelin receptor signaling: new insight into its regulatory mechanisms. J Pharmacol Sci. 2013;123(2):85–101. doi: 10.1254/jphs.13r02cr [DOI] [PubMed] [Google Scholar]
  • 63.Mehta PK, Griendling KK. Angiotensin II cell signaling: physiological and pathological effects in the cardiovascular system. Am J Physiol Cell Physiol. 2007;292(1):C82–97. doi: 10.1152/ajpcell.00287.2006 [DOI] [PubMed] [Google Scholar]
  • 64.Shui H-A, Ka S-M, Lin J-C, Lee J-H, Jin J-S, Lin Y-F, et al. Fibronectin in blood invokes the development of focal segmental glomerulosclerosis in mouse model. Nephrol Dial Transplant. 2006;21(7):1794–802. doi: 10.1093/ndt/gfl113 [DOI] [PubMed] [Google Scholar]
  • 65.Klemis V, Ghura H, Federico G, Würfel C, Bentmann A, Gretz N, et al. Circulating fibronectin contributes to mesangial expansion in a murine model of type 1 diabetes. Kidney Int. 2017;91(6):1374–85. doi: 10.1016/j.kint.2016.12.006 [DOI] [PubMed] [Google Scholar]
  • 66.Wang Y, Krämer S, Loof T, Martini S, Kron S, Kawachi H, et al. Stimulation of soluble guanylate cyclase slows progression in anti-thy1-induced chronic glomerulosclerosis. Kidney Int. 2005;68(1):47–61. doi: 10.1111/j.1523-1755.2005.00380.x [DOI] [PubMed] [Google Scholar]
  • 67.Biro M, Munoz MA, Weninger W. Targeting Rho-GTPases in immune cell migration and inflammation. Br J Pharmacol. 2014;171(24):5491–506. doi: 10.1111/bph.12658 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Yoshimura A, Wakabayashi Y, Mori T. Cellular and molecular basis for the regulation of inflammation by TGF-beta. J Biochem. 2010;147(6):781–92. doi: 10.1093/jb/mvq043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Rossol M, Pierer M, Raulien N, Quandt D, Meusch U, Rothe K, et al. Extracellular Ca2+ is a danger signal activating the NLRP3 inflammasome through G protein-coupled calcium sensing receptors. Nat Commun. 2012;3:1329. doi: 10.1038/ncomms2339 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Sharma S, Smyth B. From Proteinuria to fibrosis: an update on pathophysiology and treatment options. Kidney Blood Press Res. 2021;46(4):411–20. doi: 10.1159/000516911 [DOI] [PubMed] [Google Scholar]
  • 71.Ruggenenti P, Perna A, Remuzzi G, GISEN Group Investigators. Retarding progression of chronic renal disease: the neglected issue of residual proteinuria. Kidney Int. 2003;63(6):2254–61. doi: 10.1046/j.1523-1755.2003.00033.x [DOI] [PubMed] [Google Scholar]
  • 72.Liu D, Wen Y, Tang T-T, Lv L-L, Tang R-N, Liu H, et al. Megalin/Cubulin-Lysosome-mediated Albumin Reabsorption Is Involved in the Tubular Cell Activation of NLRP3 Inflammasome and Tubulointerstitial Inflammation. J Biol Chem. 2015;290(29):18018–28. doi: 10.1074/jbc.M115.662064 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Böttinger EP. TGF-beta in renal injury and disease. Semin Nephrol. 2007;27(3):309–20. doi: 10.1016/j.semnephrol.2007.02.009 [DOI] [PubMed] [Google Scholar]
  • 74.Maack T, Suzuki M, Almeida FA, Nussenzveig D, Scarborough RM, McEnroe GA, et al. Physiological role of silent receptors of atrial natriuretic factor. Science. 1987;238(4827):675–8. doi: 10.1126/science.2823385 [DOI] [PubMed] [Google Scholar]
  • 75.Chusho H, Tamura N, Ogawa Y, Yasoda A, Suda M, Miyazawa T, et al. Dwarfism and early death in mice lacking C-type natriuretic peptide. Proc Natl Acad Sci U S A. 2001;98(7):4016–21. doi: 10.1073/pnas.071389098 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Tamura N, Doolittle LK, Hammer RE, Shelton JM, Richardson JA, Garbers DL. Critical roles of the guanylyl cyclase B receptor in endochondral ossification and development of female reproductive organs. Proc Natl Acad Sci U S A. 2004;101(49):17300–5. doi: 10.1073/pnas.0407894101 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Tsuji T, Kunieda T. A loss-of-function mutation in natriuretic peptide receptor 2 (Npr2) gene is responsible for disproportionate dwarfism in cn/cn mouse. J Biol Chem. 2005;280(14):14288–92. doi: 10.1074/jbc.C500024200 [DOI] [PubMed] [Google Scholar]
  • 78.Packer M, Claggett B, Lefkowitz MP, McMurray JJV, Rouleau JL, Solomon SD, et al. Effect of neprilysin inhibition on renal function in patients with type 2 diabetes and chronic heart failure who are receiving target doses of inhibitors of the renin-angiotensin system: a secondary analysis of the PARADIGM-HF trial. Lancet Diabetes Endocrinol. 2018;6(7):547–54. doi: 10.1016/S2213-8587(18)30100-1 [DOI] [PubMed] [Google Scholar]
  • 79.Mc Causland FR, Lefkowitz MP, Claggett B, Anavekar NS, Senni M, Gori M, et al. Angiotensin-neprilysin inhibition and renal outcomes in heart failure with preserved ejection fraction. Circulation. 2020;142(13):1236–45. doi: 10.1161/CIRCULATIONAHA.120.047643 [DOI] [PubMed] [Google Scholar]
  • 80.Haynes R, Judge PK, Staplin N, Herrington WG, Storey BC, Bethel A, et al. Effects of sacubitril/valsartan versus irbesartan in patients with chronic kidney disease. Circulation. 2018;138(15):1505–14. doi: 10.1161/CIRCULATIONAHA.118.034818 [DOI] [PubMed] [Google Scholar]
  • 81.Choi MR, Fernández BE. Protective renal effects of atrial natriuretic peptide: where are we now?. Front Physiol. 2021;12:680213. doi: 10.3389/fphys.2021.680213 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Durand-Arczynska W, Marmy N, Durand J. Caldesmon, calponin and alpha-smooth muscle actin expression in subcultured smooth muscle cells from human airways. Histochemistry. 1993;100(6):465–71. doi: 10.1007/BF00267827 [DOI] [PubMed] [Google Scholar]
  • 83.Lessard JL. Two monoclonal antibodies to actin: one muscle selective and one generally reactive. Cell Motil Cytoskeleton. 1988;10(3):349–62. doi: 10.1002/cm.970100302 [DOI] [PubMed] [Google Scholar]
  • 84.Wong DWL, Yiu WH, Chan KW, Li Y, Li B, Lok SWY, et al. Activated renal tubular Wnt/beta-catenin signaling triggers renal inflammation during overload proteinuria. Kidney Int. 2018;93(6):1367–83. Epub 2018/04/02. doi: 10.1016/j.kint.2017.12.017 PubMed ; PubMed Central PMCID: PMCPMC5967994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wang L, Sha Y, Bai J, Eisner W, Sparks MA, Buckley AF, et al. Podocyte-specific knockout of cyclooxygenase 2 exacerbates diabetic kidney disease. Am J Physiol Renal Physiol. 2017;313(2):F430–9. doi: 10.1152/ajprenal.00614.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Whitesall SE, Hoff JB, Vollmer AP, D’Alecy LG. Comparison of simultaneous measurement of mouse systolic arterial blood pressure by radiotelemetry and tail-cuff methods. Am J Physiol Heart Circ Physiol. 2004;286(6):H2408–15. doi: 10.1152/ajpheart.01089.2003 [DOI] [PubMed] [Google Scholar]
  • 87.Shigehara T, Zaragoza C, Kitiyakara C, Takahashi H, Lu H, Moeller M, et al. Inducible podocyte-specific gene expression in transgenic mice. J Am Soc Nephrol. 2003;14(8):1998–2003. doi: 10.1681/ASN.V1481998 [DOI] [PubMed] [Google Scholar]
  • 88.Schwartz EJ, Cara A, Snoeck H, Ross MD, Sunamoto M, Reiser J, et al. Human immunodeficiency virus-1 induces loss of contact inhibition in podocytes. J Am Soc Nephrol. 2001;12(8):1677–84. doi: 10.1681/ASN.V1281677 [DOI] [PubMed] [Google Scholar]

Decision Letter 0

Michael Bader

10 Sep 2024

PONE-D-24-36618Podocyte specific knockout of the natriuretic peptide clearance receptor is podocyte protective in focal segmental glomerulosclerosisPLOS ONE

Dear Dr. Spurney,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process. Please submit your revised manuscript by Oct 25 2024 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org . When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols . Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols .

We look forward to receiving your revised manuscript.

Kind regards,

Michael Bader

Academic Editor

PLOS ONE

Journal requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2.  We noticed you have some minor occurrence of overlapping text with the following previous publication(s), which needs to be addressed:

https://pubmed.ncbi.nlm.nih.gov/34755480/

In your revision ensure you cite all your sources (including your own works), and quote or rephrase any duplicated text outside the methods section. Further consideration is dependent on these concerns being addressed.

3.  Please note that funding information should not appear in any section or other areas of your manuscript. We will only publish funding information present in the Funding Statement section of the online submission form. Please remove any funding-related text from the manuscript.

4. We note that the grant information you provided in the ‘Funding Information’ and ‘Financial Disclosure’ sections do not match. When you resubmit, please ensure that you provide the correct grant numbers for the awards you received for your study in the ‘Funding Information’ section.

5. Thank you for stating the following financial disclosure:

“1. R21 TR004257-01 from the National Institutes of Health.

2.  BX002984 from the Veterans Administration Merit Review Program.

3.  W81XWH-19-1-0314 from the United States Department of Defense.”

Please state what role the funders took in the study. If the funders had no role, please state: "The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript." If this statement is not correct you must amend it as needed. Please include this amended Role of Funder statement in your cover letter; we will change the online submission form on your behalf.

6. We note that your Data Availability Statement is currently as follows: All relevant data are within the manuscript and its Supporting Information files.

Please confirm at this time whether or not your submission contains all raw data required to replicate the results of your study. Authors must share the “minimal data set” for their submission. PLOS defines the minimal data set to consist of the data required to replicate all study findings reported in the article, as well as related metadata and methods (https://journals.plos.org/plosone/s/data-availability#loc-minimal-data-set-definition). For example, authors should submit the following data: - The values behind the means, standard deviations and other measures reported; - The values used to build graphs; - The points extracted from images for analysis. Authors do not need to submit their entire data set if only a portion of the data was used in the reported study. If your submission does not contain these data, please either upload them as Supporting Information files or deposit them to a stable, public repository and provide us with the relevant URLs, DOIs, or accession numbers. For a list of recommended repositories, please see https://journals.plos.org/plosone/s/recommended-repositories. If there are ethical or legal restrictions on sharing a de-identified data set, please explain them in detail (e.g., data contain potentially sensitive information, data are owned by a third-party organization, etc.) and who has imposed them (e.g., an ethics committee). Please also provide contact information for a data access committee, ethics committee, or other institutional body to which data requests may be sent. If data are owned by a third party, please indicate how others may request data access.

7. When completing the data availability statement of the submission form, you indicated that you will make your data available on acceptance. We strongly recommend all authors decide on a data sharing plan before acceptance, as the process can be lengthy and hold up publication timelines. Please note that, though access restrictions are acceptable now, your entire data will need to be made freely accessible if your manuscript is accepted for publication. This policy applies to all data except where public deposition would breach compliance with the protocol approved by your research ethics board. If you are unable to adhere to our open data policy, please kindly revise your statement to explain your reasoning and we will seek the editor's input on an exemption. Please be assured that, once you have provided your new statement, the assessment of your exemption will not hold up the peer review process.

8. Please include your full ethics statement in the ‘Methods’ section of your manuscript file. In your statement, please include the full name of the IRB or ethics committee who approved or waived your study, as well as whether or not you obtained informed written or verbal consent. If consent was waived for your study, please include this information in your statement as well.

9. Please include captions for your Supporting Information files at the end of your manuscript, and update any in-text citations to match accordingly. Please see our Supporting Information guidelines for more information: http://journals.plos.org/plosone/s/supporting-information.

10. PLOS ONE now requires that authors provide the original uncropped and unadjusted images underlying all blot or gel results reported in a submission’s figures or Supporting Information files. This policy and the journal’s other requirements for blot/gel reporting and figure preparation are described in detail at https://journals.plos.org/plosone/s/figures#loc-blot-and-gel-reporting-requirements and https://journals.plos.org/plosone/s/figures#loc-preparing-figures-from-image-files. When you submit your revised manuscript, please ensure that your figures adhere fully to these guidelines and provide the original underlying images for all blot or gel data reported in your submission. See the following link for instructions on providing the original image data: https://journals.plos.org/plosone/s/figures#loc-original-images-for-blots-and-gels. In your cover letter, please note whether your blot/gel image data are in Supporting Information or posted at a public data repository, provide the repository URL if relevant, and provide specific details as to which raw blot/gel images, if any, are not available. Email us at plosone@plos.org if you have any questions.

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Partly

Reviewer #2: Partly

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: Comments to “Podocyte specific knockout of the natriuretic peptide clearance receptor is podocyte protective in focal segmental glomerulosclerosis” from Liming Wang, Yuping Tang, Anne F. Buckley and Robert F. Spurney

This experimental study addresses the original and clinically relevant question whether deletion (KO) of the natriuretic peptide receptor C (NPR-C) in podocytes augments the renoprotective effects of natriuretic peptides. To this aim, the authors generated mice with deletion of NPRC in podocytes and studied them in an experimental model of glomerulosclerosis. The study focusses on C-type natriuretic peptide (CNP). An explanation why ANP and BNP were not considered as mediators of the protective effects of NPRC KO is not provided.

The results are potentially interesting but the quality of the experiments, including the quality of the illustrating figures, are too low to evaluate validity (specific criticisms are listed below). This especially concerns the illustrations of immunohistochemistry (stainings are diffuse, negative controls and scale bars are missing). Mechanistic insights to the antiapoptotic and antifibrotic effects of podocyte NPRC deletion are not provided. Moreover, the authors did not consider the possibility that the “protection” of NPRC-deficient podocytes was partly mediated by inhibition of the Gi/Gq-coupling functions of this receptor (doi: 10.1038/s41392-023-01560-y; and many other references). In other words, a clear demonstration whether NPRC KO was protective due to augmented NP/NPRB (or NPRA)/cGMP signalling and/or due to diminished NPRC/G-protein signalling is missing. This limitation must be addressed or at least carefully discussed.

Specific Comments:

- Introduction (page 3): “1. Podocytes express NPRC, NPRB and NPRC [13-17]”. Please correct the redundancy in this sentence.

- Nomenclature: for many good reasons, the IUPHAR recommended to name NPRA and NPRB as GC-A and GC-B. At least you should clarify this nomenclature.

- Introduction (page 4): “1. NPs potently stimulated cGMP generation and inhibited podocyte apoptosis in cultured podocytes[17], and 2. Pharmacological blockade of NPs in vivo enhanced urinary cGMP excretion and significantly reduced albuminuria in a mouse model of FSGS[17].” These two sentences obviously contradict each other. Please correct them or discuss appropriately.

- Throughout the manuscript there are numerous grammatical mistakes and incomplete sentences, which leaves the reviewer with the impression of quick superficial writings (for instance on pages 1 and 2 of results section: detecteded, anibody, exression, nomral, podocytres, …)

- Figure 1:

o The quality of the images in Figure 1A is very low and must be markedly improved.

o Figures 1B and C illustrate “statements” (CNP is produced by podocytes and binds to…; NPRC clears CNP from the circulation) which were not experimentally addressed by the present studies. If these statements/schemes are based on published data, then please refer those in the legend. As far as this reviewer knows, nobody has previously shown that “NPRC clears CNP from the circulation”).

o Figure 1E: How is the expression of NPRB and NPRC in these cells? Is it correct that CNP barely increased cGMP contents of cultured control podocytes but raised the cGMP contents of NPR-C KO podocytes by more than 50-fold? This is a surprisingly huge difference. It suggests that in presence of NPR-C, CNP/NPRB/cGMP signalling is almost fully blocked. Why? As compared: how are the cGMP responses of control and KD podocytes to ANP and/or BNP?

o Figure 1G: results are unclear and possibly incomplete. Please show the % apoptosis in control and KO cells in Serum and Serum-free medium. Is there a difference in serum-containing medium between genotypes? The increased apoptosis is interesting and should be confirmed with additional techniques and complemented with mechanistic insights.

- Figure 2:

o How is the protein level of NPR-C in glomeruli from both genotypes? Is NPR-C expression preserved in other types of cells? (was NPR-C expression in rest of kidney, after separation of glomeruli, preserved?)

- Figure 4:

o Figure 4C: The y-axis is named number of mice with Glomerulosclerosis. The legend shows normal, mild, moderate and severe sclerosis. Please label the y-axis accordingly (staging of glomerulosclerosis, histology of glomeruli?). It would help understandings to present the data in %, as is described in corresponding results section. (“~86% of the WT controls have no glomerulosclerosis and ~14 % mild glomerulosclerosis”). The present Figure is totally unclear because it does not relate the “depicted numbers” to the total number of study mice.

o Figure 4D-F: The columns “Absent” and “Present” in the diagram are redundant. Please show only the column “Present”. Why did you interchange black/white in Fig. E?

o Does podocyte apoptosis induce tubulointerstitial inflammation? Where is the link between these two observations? Figure 4E must illustrate in clear way how inflammation was measured.

- Figure 5:

o The mRNA and protein results are very discrepant. The mRNA expression levels of alpha-SMA and Fibronectin are only slightly impacted by genotypes and conditions (Figure 5A-D). The Western blots instead show huge inductions in WT mice and almost full inhibition of this induction in the KO (Figures 5E-H). How do you explain this mismatch between the regulation of mRNA and protein levels?

- Table S2:

o As mentioned in the discussion, NPs inhibit the master regulator of fibrosis, TGF-beta, by phosphorylating SMADs on an inhibitory site. Did you analyse SMAD phosphorylation? Is the inhibitory phosphorylation increased in the KO mice? Do NPRC-deficient podocytes express less proinflammatory and profibrotic molecules?

- Please discuss appropriately the discrepant results from Figures 3 (marked Albuminuria) and 7 (Cystatin C levels barely affected)

- Methods: It is difficult to understand how the diverse transgenic mouse models were generated. It could help to include a scheme of the mice breedings.

Reviewer #2: The present work deals with the role of the natriuretic peptide receptor C (NPRC), which has a functional significance as a clearance receptor for the natriuretic peptides ANP, BNP and CNP, in podocytes. In the first part of the paper, the authors show that cultured murine podocytes express CNP and that knockdown of NPRC is associated with higher CNP levels in the medium and attenuated apoptosis induced by serum deprivation. The authors conclude that blockade or inactivation of NPRC in podocytes may have a protective effect in glomerular injury models. They therefore exposed mice with podocyte-specific deletion of NPRC to a corresponding damage model (FSGS model). Indeed, some glomerular and interstitial damage markers are ameliorated in NPRC KO mice, supporting the authors' hypothesis.

The study is the logical continuation of earlier studies by the authors with pharmacological blockade of the NPRC. However, there are some issues to consider.

1. The two parts of the study, in vitro and in vivo, are linked by the fact that the NPRC was genetically inactivated in both. Otherwise, the results of the in vitro studies are not taken up in the in vivo part. Does the local production of CNP in podocytes play also a role in vivo in terms of an autocrine/paracrine effect? Is CNP regulated in podocytes in the animal model used? Or are the observed protective effects of NPRC deletion due to an enhanced effect of ANP / BNP via the NPRA? It is understandable that this question is very difficult to answer experimentally, but it should at least be discussed in more detail.

2. Line 6, results: Reference no. 23 is given as a reference for the expression of NPRB in podocytes. This should be checked, as the paper mentioned deals with the expression of CNP in the kidney, but not with NPRB. Other studies cited as evidence for the expression of NPRB in podocytes also show very strong expression of NPRA (receptor for ANP and BNP) and NPRC, but at best very weak expression of NPRB in podocytes (references 14 and 16). On the contrary, a recent study using in situ hybridization showed the expression of NPRB in glomerular endothelial cells and mesangial cells, but not in podocytes (Heinl, ES et al.; Pflugers Archiv 2023, 475(3): 343-360). Taking into account the results of these studies, in particular the strongly dominant expression of NPRA in podocytes, the influence of NPRC knockdown on the effects of ANP or BNP in cultured podocytes (cGMP, apoptosis) should be investigated.

3. Fig. 1A: CNP immunofluorescence staining, at least in the present PDF, is not of sufficient quality to detect co-staining in podocytes. The quality of the image should be improved. It is also noted that "CNP was widely expressed in multiple cell types in the kidney". Please show overview images that reflect this expression.

4. Fig. 1E: It is clearly shown that the effect of CNP on cGMP levels is significantly enhanced in NPRC knockdown cells compared to control cells. It is not clear from Fig. 1E whether CNP has a significant effect at all in control cells. Does cGMP increase in control cells under CNP or is the effect completely abolished in the presence of NPRC.

5. The results of the in vitro experiments are compatible with the conclusion that the endogenous production of CNP in podocytes has a protective effect and that this is mediated via the NPRB. To further substantiate this, the NPRB could be knocked down in the podocyte cell culture used, with and without simultaneous NPRC inactivation, and the effect on apoptosis investigated. Overall, it should at least be discussed that there are a variety of studies showing that the NPRC is not only a clearance receptor, but also a signaling receptor that can be activated by CNP (for example: Bubb KJ, et. al.; Circulation. 2019 ;139(13): 1612-1628.; Moyes AJ, Hobbs AJ. Int J Mol Sci. 2019 ;20(9): 2281).

6. Fig. 4: Please show absolute values and not percentages.

7. Fig. 5G: The arrow for fibronectin does not point to the dominant band of the Western blot. Which is the correct band? The molecular weight or a standard should be shown.

8. First page of introduction, last line: It should read "NPRA, NPRB and NPRC" instead of "NPRC, NPRB and NPRC".

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean? ). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy .

Reviewer #1: No

Reviewer #2: No

**********

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/ . PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org . Please note that Supporting Information files do not need this step.

PLoS One. 2025 Mar 10;20(3):e0319424. doi: 10.1371/journal.pone.0319424.r003

Author response to Decision Letter 0


21 Nov 2024

Reviewer #1:

1. Histologic studies: Immunohistochemistry staining is diffuse and lacks negative controls.

We had significant difficulty locating antibodies for the immunohistochemistry studies, and were unable to locate antibodies for mouse kidney. As a result, we attempted to stain human kidney specimens with several antibodies and the current figure was the result. The antibody we used stained multiple cell types in the kidney the kidney consistent with published single cell transcriptomic studies (for example see reference [1]) Little staining was detected in the isotype control antibody panels. We agree that the images a suboptimal and it is also possible that the diffuse staining in the tope panels of Figure 1A could be non-specific. As a result, we are removing Figure 1A from the manuscript.

2. Scale bars are missing:

We have added scale bars to the current histology pictures figures.

3. Alternative considerations: Did not consider that the “protection” of NPRC-deficient podocytes was partly mediated by inhibition of the Gi/Gq-coupling functions of this receptor.

We are aware of alternative signaling pathways linked to NPRC and we discussed this possibility in a previous publication examining the effects of pharmacologic NPRC blockade in a mouse model of FSGS [2]. We apologize for not include this possibility in the current publication and we have amended the manuscript to address this possibility in the revised manuscript.

4. Nomenclature: for many good reasons, the IUPHAR recommended to name NPRA and NPRB as GC-A and GC-B. At least you should clarify this nomenclature.

We have acknowledged the alternate designations for NPRA and NPRB in the text of the manuscript.

5 Introduction: Contradictory sentences: “1. NPs potently stimulated cGMP generation and inhibited podocyte apoptosis in cultured podocytes[17], and 2. Pharmacological blockade of NPs in vivo enhanced urinary cGMP excretion and significantly reduced albuminuria in a mouse model of FSGS[17].” These two sentences obviously contradict each other. Please correct them or discuss appropriately.

We apologize for the error, and we have corrected the mistake as outlined below.

The corrected statement is shown below:

NPs potently stimulated cGMP generation and inhibited podocyte apoptosis in cultured podocytes[17], and 2. Pharmacological blockade of NPRC in vivo enhanced urinary cGMP excretion and significantly reduced albuminuria in a mouse model of FSGS[17].” These two sentences obviously contradict each other. Please correct them or discuss appropriately.

6. Figure 1A: The quality of the images in Figure 1A is very low and must be markedly improved.

We agree that the Immunohistochemistry studies are suboptimal. See response #1 above.

6. Figures 1B and C illustrate “statements” (CNP is produced by podocytes and binds to…; NPRC clears CNP from the circulation) which were not experimentally addressed by the present studies. If these statements/schemes are based on published data, then please refer those in the legend. As far as this reviewer knows, nobody has previously shown that “NPRC clears CNP from the circulation”).

The natriuretic peptide (NP) clearance receptor (NPRC) binds ANP, BNP, and CNP with a high affinity in the low picomolar range [3-6] and either pharmacologic blockade or KO of NPR-C suggests that a major function of NPRC is to clear NPs from the circulation [7, 8]. The negative regulatory effects of NPRC on the functions of CNP have been demonstrated in bone using mouse models. In this regard, mutations that disrupt the function of genes encoding CNP or NPRB (CNP receptor) causes dwarfism [9-11]. In contrast, disruption of the gene encoding the clearance receptor (NPRC) cause bone overgrowth [8]. These data suggest that: 1. A major function of CNP is binding to NPRB and stimulating bone growth, and 2. the stimulatory effects of CNP on bone growth are augmented by decreased clearance of CNP from the circulation by NPRC.

7. How is the expression of NPRB and NPRC in these cells?

We have provided data on expression of NPRA, NPRB and NPRC in Figure 2. The major findings were: 1. NPRC expression was effectively decreased in KD cells, 2. NPRC is robustly expressed in podocytes compared to either NPRA or NPRB, and 3. NPRB expression is downregulated in NPRC KO mice.

8. Is it correct that CNP barely increased cGMP contents of cultured control podocytes but raised the cGMP contents of NPR-C KO podocytes by more than 50-fold? This is a surprisingly huge difference. It suggests that in presence of NPR-C, CNP/NPRB/cGMP signaling is almost fully blocked. Why?

It is correct that CNP induced cGMP generation is significantly blunted by the high expression levels of both NPRC [1, 12, 13] and proteolytic cleavage of NPs by neprilysin [2, 14, 15]. Moreover, the 0.01 nM concentration of CNP is at the lower limit of NPRB binding affinity [5, 6, 16], which severly limits the response to CNP at the 0.01 nM concentration (Figure 1E). As a result, calculating the fold increase in cGMP generation using a number close to zero results in a very large fold change. In contrast, the higher concentrations of CNP are more effectively bound by NPRB (GC-B) and results in more robust cGMP response in, although the response is likely tempered by the by both the clearance receptor (NPRC) and peptide cleavage by neprilysin (both are expressed in podocytes [2]).

9. Figure 1E: As compared: how are the cGMP responses of control and KD podocytes to , and/or BNP?

We previously measured cGMP generation using 1µM concentrations of ANP and CMP. In these studies, ANP stimulated cAMP to a greater extent than CNP but the difference was not statistically different [2]. In the current study (Figure 2A), we saw the same pattern. ANP stimulated a greater increase in cGMP generation compared to CNP, but the differences were not statistically different at the 1.0 nM or 10 nM concentrations. In addition, the same pattern was observed in NPRC KD cells, although cGMP response was enhanced compared to cells expressing NPRC. See Figure 2A for current results.

10. Figure 1G: Results are unclear and possibly incomplete. Please show the % apoptosis in control and KO cells in Serum and Serum-free medium. Is there a difference in serum-containing medium between genotypes?

As described in the initial submission, we combined serum treated control and KD podocyte data because little apoptosis was observed in both groups. In the revised manuscript, we have repeated the experiments and separated both the serum and serum free data for KD and control podocytes. In addition, we have increased the duration of the serum deprivation to enhance the apoptotic effect to increase the apoptotic response and, in turn, provide a greater range to enhance sensitivity for detecting differences. In the current study (Figure 1F), KD of NPRC reduced apoptosis induced by serum deprivation, and this beneficial effect of NPRC KD was partially attenuated by pharmacologic blockade of NPRB with pharmacologic blockade pf NPRB with P19 [17, 18]. There was no statistically significant difference in apoptosis between control and KD cells treated with serum. These data are consistent with the notion that beneficial effects of NPRC KO on the apoptotic response was mediated, at least part, by activation of NPRB by CNP.

11. Figure 1G: The increased apoptosis is interesting and should be confirmed with additional techniques and complemented with mechanistic insight.

Serum deprivation has been utilized for several decades to induce cell death by the removal of pro-survival growth factors [19]. Moreover, our lab and others have used serum deprivation to induce apoptosis in podocytes [2, 20]. In response to your comment, the mechanisms causing cell death has expanded to include multiple distinct pathways [21]. In addition, there is overlap between of the pathways promoting cell death. For example, autophagy often progresses to apoptosis [21, 22] and if apoptotic cells are not efficiently cleared, the cells undergo programed cell necrosis [21-23]. The methodology used in our study (annexin V staining) is commonly used to detect apoptosis and, given the complexity of investigating types of cells death and the current focus of this manuscript, we believe the studies are beyond the focus of this manuscript.

12. Figure 2: How is the protein level of NPR-C in glomeruli from both genotypes? Is NPR-C expression preserved in other types of cells? (was NPR-C expression in rest of kidney, after separation of glomeruli, preserved?)

As discussed in the methods section, the Cre mouse was obtained from Jackson labs (model No. 008205) and permits inducible expression of Cre recombinase specifically in podocytes. The podocyte specificity of the system was documented in the initial publication [24] and this model has been used in multiple manuscripts since the initial paper was published. Figure 3A&B and supplementary figure S2 demonstrate that expression of Cre recombinase under the regulation of the podocyte specific podocin promoter in tdTomato mice (see methods) causes expression of green fluorescence in a pattern consistent with expression of Cre recombinase specifically in podocytes. In mice containing 2 “floxed” alleles of NPRC, this same promoter system specifically deletes NPRC in podocytes. In addition, we demonstrated that levels of NPRC were significantly decreased in the glomerular preparations by both quantitative RT-PCR (figure 2C) and immunoblotting (figure 2D & 2E) despite expression of NPRC in multiple cell types in the kidney [1, 12, 25]. Taken together with a large literature suggesting specificity of this approach to KO of podocyte proteins, these data suggest that podocyte specific KO of NPRC was successful.

13. Figure 4: The y-axis is named number of mice with Glomerulosclerosis. The legend shows normal, mild, moderate and severe sclerosis. Please label the y-axis accordingly (staging of glomerulosclerosis, histology of glomeruli?). It would help understandings to present the data in %, as is described in corresponding results section. (“~86% of the WT controls have no glomerulosclerosis and ~14 % mild glomerulosclerosis”). The present Figure is totally unclear because it does not relate the “depicted numbers” to the total number of study mice. Figure 4D-F: The columns “Absent” and “Present” in the diagram are redundant. Please show only the column “Present”.

The percentage of mice with glomerulosclerosis (GS) relates to number of mice with (absent) or without (Present) evidence of GS. For example, in Figure 4C, the TG wild type group has 3 mice without GS, 5 mice with mild GS, 2 mice with moderate GS and 11 mice with severe GS. The total number of mice is 21. As a result, there are 18 mice with GS (5+2+11) divided by 21 mice without GS, which is approximately ~85.7% of TG wild type mice with GS.

To simplify Figure 4, we have eliminated Figure 4C from the manuscript and reported the percentage of mice with or without GS; however, we have retained Figure 4C in the Supporting Data to provide the exact numbers of mice with mild, moderate and severe GS as reported by our pathologist (Dr. Buckley). We decided to present the graphs of the histopathologic findings as the percentage of mice with the specified abnormality to permit a more effective comparison of the differences between the experimental groups in studies with an imbalance in the number of mice in each group; however, the statistical evaluation was performed using the number of mice with the specified histologic abnormality.

14. Figure 4E: Why did you interchange black/white in Fig. E?

We have corrected the black/white problem in labeling the percentages.

15. Figure 4E: Does podocyte apoptosis induce tubulointerstitial inflammation? Where is the link between these two observations?

Podocyte damage/cell death disrupts the glomerular filtration barrier and promotes high levels of proteinuria. As discussed in the text of the manuscript, the level of proteinuria may contribute to tubulointerstitial inflammation. Filtered proteins are resorbed by the proximal tubules and processed by lysosomes and endoplasmic reticulum [26]. In the setting of heavy proteinuria, these intracellular pathways are stressed [27], which results in the secretion of cytokines that attract and activate inflammatory cells [28] and produce profibrotic mediators such as TGF-beta [29].

16. Figure4E: Figure 4E must illustrate in clear way how inflammation was measured.

The pathologist evaluating the renal histopathology provided a detailed descript of the methodology for assessing tubulointerstitial inflammation. Please see the Methods Section for details.

17. Figure 5: The mRNA and protein results are very discrepant. The mRNA expression levels of alpha- SMA and Fibronectin are only slightly impacted by genotypes and conditions (Figure 5A-D). The Western blots instead show huge inductions in WT mice and almost full inhibition of this induction in the KO (Figures 5E-H). How do you explain this mismatch between the regulation of mRNA and protein levels?

We also noticed the discrepancy in the mRNA expression and protein levels in the glomerular preparations. Review of the literature suggests that fibronectin accumulates in the glomerulus through both local synthesis and by deposition from the circulation [30, 31]. This was demonstrated by a liver specific fibronectin knockout to decrease circulating fibronectin [31]. In these KO mice, there is a decrease in both mesangial expansion and fibronectin accumulation the kidneys of diabetic mice. In addition, the major source of fibronectin in early stages of FSGS is from the circulation with deposition of glomerular fibronectin occurring later in the disease process. We suspect that the discrepancy in the mRNA expression and protein levels is related to greater deposition of fibronectin from the circulation in damaged glomeruli compared to glomeruli with less severe glomerular injury. This possibility has been mentioned in the discussion section.

18. As mentioned in the discussion, NPs inhibit the master regulator of fibrosis, TGF-beta, by phosphorylating SMADs on an inhibitory site. Did you analyze SMAD phosphorylation? Is the inhibitory phosphorylation increased in the KO mice? Do NPRC-deficient podocytes express less proinflammatory and profibrotic molecules?

TGF-beta activates its receptor, which phosphorylates Smad3 on Ser423 and Ser425 [32].. Phosphorylated Smad3 combines with Smad4 to form a Smad complex,which then translocates into the nucleus to initiate mRNA transcription associate with inflammation and collagen formation [32].. ANP stimulates cGMP generation and actives cGMP kinase 1 (cGK1), which then  phosphorylates Smad3 on Ser309 and on Thr388, and inhibits the nuclear translocation of the Smad complex and , in turn, fibrosis [32]. 00042.2016). Despite an extensive search, we were unable to locate antibodies to phospho-SMAD3 Ser309 or phospho-SMAD3 Thr388.

There are antibodies for phosphor-Smad3 on Ser423 and Ser425, and we attempted to determine if these Smad phosphoryaltion sites were affected in our KO mice but were unable to detect a signal. It is difficult to interpret these results because exploring cell signaling in animal models is complicated by: 1. Changes in modifications to cell signaling molecules involved in the response (for example phosphorylation and dephosphorylation in vivo, and repeated freeze-thaw cycles of stored samples ) and 2. The time point chosen to harvest tissues (late versus early in the fibrotic response). We did not screen for additional proinflammatory or profibrotic molecules.

20. Proteinuria versus kidney function: Please discuss appropriately the discrepant results from Figures 3 (marked Albuminuria) and 7 Cystatin C levels barely affected)

It is common to have a large disparity in proteinuria and renal function in both animals and humans, especially in the early stages of glomerular diseases. The amount of proteinuria is dependent on the integrity of the filtration barrier, and podocytes play a key role in maintaining permselectivity. In contras

Attachment

Submitted filename: Respomse-to-Reviewers.pdf

pone.0319424.s002.pdf (107.4KB, pdf)

Decision Letter 1

Michael Bader

6 Dec 2024

PONE-D-24-36618R1Podocyte specific knockout of the natriuretic peptide clearance receptor is podocyte protective in focal segmental glomerulosclerosisPLOS ONE

Dear Dr. Spurney,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

Please submit your revised manuscript by Jan 20 2025 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org . When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols . Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols .

We look forward to receiving your revised manuscript.

Kind regards,

Michael Bader

Academic Editor

PLOS ONE

Journal Requirements:

Please review your reference list to ensure that it is complete and correct. If you have cited papers that have been retracted, please include the rationale for doing so in the manuscript text, or remove these references and replace them with relevant current references. Any changes to the reference list should be mentioned in the rebuttal letter that accompanies your revised manuscript. If you need to cite a retracted article, indicate the article’s retracted status in the References list and also include a citation and full reference for the retraction notice.

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #2: (No Response)

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #2: Partly

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #2: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #2: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #2: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #2: The authors have addressed most of my criticisms and suggestions and have significantly improved the manuscript. However, there are still points that need to be clarified:

1. As suggested, experiments were carried out on immortalized podocytes to examine the relevance of CNP signaling via NPRB. For this purpose, the substance P19 was used (Figure 1G). The effect of P19 in KD cells is low and the significance is very limited with a test number of n=4. This could easily be corrected by additional experiments. Furthermore, the concentration of P19 used is not indicated in the manuscript. This is particularly important because the affinity of P19 for NPRA and NPRB does not differ greatly, and the greatest affinity is actually for NPRC. This information can be found in reference 26 and should at least be discussed with all its consequences. A knockdown of NPRB would be the better method here.

2. The experiments comparing the effects of ANP and CNP on cGMP and apoptosis are very helpful. They clearly show that ANP exerts a protective effect on podocytes even without simultaneous inactivation of NPRC. This fits very well with in vivo experiments by other groups. However, the finding that a protective effect of CNP is only present when NPRC is inactivated calls into question the in vivo relevance of this effect, since podocytes in vivo also express NPRC. This should be discussed.

3. As in the first version of the manuscript, the references should be carefully checked. For example, references 14 and 16 are mentioned to prove that “CNP is widely expressed in multiple cell types in mouse and human kidneys including glomerular podocytes (results, first paragraph)”. This information cannot be found in the papers. There may be a misunderstanding because “CNP” is mentioned in the gene lists. However, this is the gene of a “cyclic nucleotide phoshodiesterase” and not CNP whose gene name is NPPC. In addition, the references are used as evidence for “for the formation of cGMP in podocytes by the NPRB” (results, first paragraph). This is also not correct. Please correct me if I am wrong.

4. It should be briefly discussed that the blood pressure values show a very high variability. Although this can occasionally occur with automated tail cuff measurement and is therefore not very unusual, it significantly weakens the authors' conclusion that there are no blood pressure differences between the genotypes.

5. Legend Figure 2B: “KO mice” should be corrected to “KD cells”

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean? ). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy .

Reviewer #2: No

**********

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/ . PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org . Please note that Supporting Information files do not need this step.

PLoS One. 2025 Mar 10;20(3):e0319424. doi: 10.1371/journal.pone.0319424.r005

Author response to Decision Letter 1


13 Jan 2025

RESPONSE TO THE REVIEW

Comments to the authors:

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

- Reviewer #2: No response

- We have responded to individual comments below.

2. Is the manuscript technically sound, and do the data support the conclusions? The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

- Reviewer #2: Partly

- We have made changes in the manuscript as outlined below to address comments raised by the reviewers.

3. Has the statistical analysis been performed appropriately and rigorously?

- Reviewer #2: Yes.

4. Have the authors made all data underlying the findings in their manuscript fully available?

- Reviewer #2: Yes

5. Is the manuscript presented in an intelligible fashion and written in standard English?

- Reviewer #2: Yes

Reviewer comments to the authors:

1. As suggested, experiments were carried out on immortalized podocytes to examine the relevance of CNP signaling via NPRB. For this purpose, the substance P19 was used (Figure 1G). The effect of P19 in KD cells is low and the significance is very limited with a test number of n=4. This could easily be corrected by additional experiments. Furthermore, the concentration of P19 used is not indicated in the manuscript. This is particularly important because the affinity of P19 for NPRA and NPRB does not differ greatly, and the greatest affinity is actually for NPRC. This information can be found in reference 26 and should at least be discussed with all its consequences. A knockdown of NPRB would be the better method here.

We have performed the experiments recommended by the reviewers and, although we only used an “N” of 4, the data is statistically significant, even after correcting for multiple comparisons. We do apologize for not including the concentrations of P19 (250 nM) in the manuscript, and this data has been added to the Methods Section. As the reviewers mentioned, the affinity of P19 for NPRA and NPRB does not differ greatly, and the greatest affinity is actually for NPRC. This is an important consideration that we did not address in the revised manuscript. Based on the following reference (Peptides 26 (2005) 517–524), we suggest that binding of P19 to either NPRC or NPRA is unlikely to affect the results in the knockdown (KD) podocytes because: 1. NPRC is absent in the KD cells, 2. The dosage of P19 used in the studies (250 nM) effectively binds NPRB (Kd 15.4 nM) but binds minimally to NPRA (NPRA Kd 575 nM). In contrast to the KD cells, NPRC is expressed in the control cells, which could affect the apoptotic response by blocking natriuretic clearance and enhancing CNP levels. We posit that blockade of NPRC in the control cells by P19 (Fig. 1G) had a minimal effect on the results because P19 has high affinity for both NPRB (Kd ~15 nM) and NPRC (0.0134 nM) and, in turn, would effectively inhibit both NP receptors (NPRB and NPRC). In this scenario, the benefits of increasing CNP levels by blockade NPRC would be reduced by pharmacologic blockade of NPRB. In the current study, the net effect was no change in the apoptotic response (Fig. 1G).

NOTE: affinity in the P19 reference is expressed as pK units and can be converted to Kd as follows pK = −log KD).

2. The experiments comparing the effects of ANP and CNP on cGMP and apoptosis are very helpful. They clearly show that ANP exerts a protective effect on podocytes even without simultaneous inactivation of NPRC. This fits very well with in vivo experiments by other groups. However, the finding that a protective effect of CNP is only present when NPRC is inactivated calls into question the in vivo relevance of this effect, since podocytes in vivo also express NPRC. This should be discussed.

We agree that, under the conditions used in the experiments (cell culture), ANP is more effect than CNP at inhibiting podocyte apoptosis at equimolar concentrations of ANP and CNP (Figure 2D). Indeed, the podocyte protective actions of CNP are only seen in the NPRC KD cells. The current studies do not, however, address potential CNP secretion under in vivo conditions or changes in CNP secretion during stress. Moreover, there are multiple CNP secreting cells in kidney that may contribute to enhance CNP levels and “overcome” the negative regulatory effects of NPRC. However, based on the data presented in the manuscript, we have modified the statements about the autocrine protective actions of CNP in the text of the manuscript to acknowledge the lack of a podocyte protective effect in the cells expressing NPRC

3. As in the first version of the manuscript, the references should be carefully checked. For example, references 14 and 16 are mentioned to prove that “CNP is widely expressed in multiple cell types in mouse and human kidneys including glomerular podocytes (results, first paragraph)”. This information cannot be found in the papers. There may be a misunderstanding because “CNP” is mentioned in the gene lists. However, this is the gene of a “cyclic nucleotide phoshodiesterase” and not CNP whose gene name is NPPC. In addition, the references are used as evidence for “for the formation of cGMP in podocytes by the NPRB” (results, first paragraph). This is also not correct. Please correct me if I am wrong.

The references 14 and 16 are single cell RNA sequencing studies. To obtain results for specific mRNAs investigators need to query the transcriptomics databases that accompany the manuscripts.

Database for these results, are available at the following websites: 1. http://humphreyslab.com/SingleCell/ and 2. https://susztaklab.com/EH/park/. I have also added these weblinks to each manuscript citing single cell RNA sequencing studies in the Reference Section. I am also requesting guidance how to reference these results in the manuscript.

4. It should be briefly discussed that the blood pressure values show a very high variability. Although this can occasionally occur with automated tail cuff measurement and is therefore not very unusual, it significantly weakens the authors' conclusion that there are no blood pressure differences between the genotypes.

The tail cuff blood pressure measurement did show variability in the results, which reduces chances of detecting a difference between groups. We can only speculate on the causes, but the major finding in the BP studies is the lack of a difference in the Gq mice compared to the Gq-KO mice. This is important because a difference in BP could affect the severity of the kidney disease. The comparison in these 2 groups (GQ and Gq-KO) found very similar BP levels suggesting that a difference in BP did not affect the in vivo findings in the study.

5. Legend Figure 2B: “KO mice” should be corrected to “KD cells”

We have corrected this mistake in the text of the manuscript.

Additional issues: PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

I do not think is necessary to publish the full review process.

References

1. Kubiak-Wlekly A, Perkowska-Ptasinska A, Olejniczak P, Rochowiak A, Kaczmarek E, Durlik M, et al. The Comparison of the Podocyte Expression of Synaptopodin, CR1 and Neprilysin in Human Glomerulonephritis: Could the Expression of CR1 be Clinically Relevant? Int J Biomed Sci. 2009;5(1):28-36. PubMed PMID: 23675111; PubMed Central PMCID: PMCPMC3614758.

2. Debiec H, Guigonis V, Mougenot B, Decobert F, Haymann JP, Bensman A, et al. Antenatal membranous glomerulonephritis due to anti-neutral endopeptidase antibodies. N Engl J Med. 2002;346(26):2053-60. Epub 2002/06/28. doi: 10.1056/NEJMoa012895. PubMed PMID: 12087141.

Attachment

Submitted filename: PLOS Rebuttal Letter.docx

pone.0319424.s003.docx (21.4KB, docx)

Decision Letter 2

Michael Bader

17 Jan 2025

PONE-D-24-36618R2Podocyte specific knockout of the natriuretic peptide clearance receptor is podocyte protective in focal segmental glomerulosclerosisPLOS ONE

Dear Dr. Spurney,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the last point that should be clarified: Please make sure that the single-cell RNAseq databases you cite (refs 14 and 16) indeed show the expression of NPPC (it can not be found in https://susztaklab.com/EH/par). If you also can't find it, please cite other references or change the text.

Please submit your revised manuscript by Mar 03 2025 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org . When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols . Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols .

We look forward to receiving your revised manuscript.

Kind regards,

Michael Bader

Academic Editor

PLOS ONE

Journal Requirements:

Please review your reference list to ensure that it is complete and correct. If you have cited papers that have been retracted, please include the rationale for doing so in the manuscript text, or remove these references and replace them with relevant current references. Any changes to the reference list should be mentioned in the rebuttal letter that accompanies your revised manuscript. If you need to cite a retracted article, indicate the article’s retracted status in the References list and also include a citation and full reference for the retraction notice.

[Note: HTML markup is below. Please do not edit.]

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/ . PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org . Please note that Supporting Information files do not need this step.

PLoS One. 2025 Mar 10;20(3):e0319424. doi: 10.1371/journal.pone.0319424.r007

Author response to Decision Letter 2


22 Jan 2025

RESPONSE TO THE REVIEW

Comments to editorial staff:

I am submitting the revised manuscript entitled "Podocyte specific knockout of the natriuretic peptide clearance receptor ameliorates focal segmental glomerulosclerosis” for consideration for publication in Plos One. I am responding to the following comment:

“Please make sure that the single-cell RNAseq databases you cite (refs 14 and 16) shows expression of NPPC (it cannot be found in https://susztaklab.com/EH/par). If you also can't find it, please cite other references or change the text.”

There are 3 references (14, 16 and 50) with links to the data bases that can be searched to locate the mRNAs by either clinking on the link or copying and pasting the link into a browser. The above link for reference 14 is incorrect (https://susztaklab.com/EH/par).

The links to the correct data sets are:

- Reverence 14: https://susztaklab.com/EH/park/

- Reference 16: http://humphreyslab.com/SingleCell/

- Reference 50: http://humphreyslab.com/SingleCell/

The links in the previous submission were correct and the only changes I have made in the manuscript were to modify the sentence identifying the data bases from, “Results for specific mRNAs can be accessed at the following database” to “Results for specific mRNAs can be accessed by searching the following database”. If this format cannot be used for references, I will eliminate references 14, 16 and 50.

Comments to the authors:

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

- Reviewer #2: No response

- We have responded to individual comments below.

2. Is the manuscript technically sound, and do the data support the conclusions? The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

- Reviewer #2: Partly

- We have made changes in the manuscript as outlined below to address comments raised by the reviewers.

3. Has the statistical analysis been performed appropriately and rigorously?

- Reviewer #2: Yes.

4. Have the authors made all data underlying the findings in their manuscript fully available?

- Reviewer #2: Yes

5. Is the manuscript presented in an intelligible fashion and written in standard English?

- Reviewer #2: Yes

Reviewer comments to the authors:

1. As suggested, experiments were carried out on immortalized podocytes to examine the relevance of CNP signaling via NPRB. For this purpose, the substance P19 was used (Figure 1G). The effect of P19 in KD cells is low and the significance is very limited with a test number of n=4. This could easily be corrected by additional experiments. Furthermore, the concentration of P19 used is not indicated in the manuscript. This is particularly important because the affinity of P19 for NPRA and NPRB does not differ greatly, and the greatest affinity is actually for NPRC. This information can be found in reference 26 and should at least be discussed with all its consequences. A knockdown of NPRB would be the better method here.

We have performed the experiments recommended by the reviewers and, although we only used an “N” of 4, the data is statistically significant, even after correcting for multiple comparisons. We do apologize for not including the concentrations of P19 (250 nM) in the manuscript, and this data has been added to the Methods Section. As the reviewers mentioned, the affinity of P19 for NPRA and NPRB does not differ greatly, and the greatest affinity is actually for NPRC. This is an important consideration that we did not address in the revised manuscript. Based on the following reference (Peptides 26 (2005) 517–524), we suggest that binding of P19 to either NPRC or NPRA is unlikely to affect the results in the knockdown (KD) podocytes because: 1. NPRC is absent in the KD cells, 2. The dosage of P19 used in the studies (250 nM) effectively binds NPRB (Kd 15.4 nM) but binds minimally to NPRA (NPRA Kd 575 nM). In contrast to the KD cells, NPRC is expressed in the control cells, which could affect the apoptotic response by blocking natriuretic clearance and enhancing CNP levels. We posit that blockade of NPRC in the control cells by P19 (Fig. 1G) had a minimal effect on the results because P19 has high affinity for both NPRB (Kd ~15 nM) and NPRC (0.0134 nM) and, in turn, would effectively inhibit both NP receptors (NPRB and NPRC). In this scenario, the benefits of increasing CNP levels by blockade NPRC would be reduced by pharmacologic blockade of NPRB. In the current study, the net effect was no change in the apoptotic response (Fig. 1G).

NOTE: affinity in the P19 reference is expressed as pK units and can be converted to Kd as follows pK = −log KD).

2. The experiments comparing the effects of ANP and CNP on cGMP and apoptosis are very helpful. They clearly show that ANP exerts a protective effect on podocytes even without simultaneous inactivation of NPRC. This fits very well with in vivo experiments by other groups. However, the finding that a protective effect of CNP is only present when NPRC is inactivated calls into question the in vivo relevance of this effect, since podocytes in vivo also express NPRC. This should be discussed.

We agree that, under the conditions used in the experiments (cell culture), ANP is more effect than CNP at inhibiting podocyte apoptosis at equimolar concentrations of ANP and CNP (Figure 2D). Indeed, the podocyte protective actions of CNP are only seen in the NPRC KD cells. The current studies do not, however, address potential CNP secretion under in vivo conditions or changes in CNP secretion during stress. Moreover, there are multiple CNP secreting cells in kidney that may contribute to enhance CNP levels and “overcome” the negative regulatory effects of NPRC. However, based on the data presented in the manuscript, we have modified the statements about the autocrine protective actions of CNP in the text of the manuscript to acknowledge the lack of a podocyte protective effect in the cells expressing NPRC

3. As in the first version of the manuscript, the references should be carefully checked. For example, references 14 and 16 are mentioned to prove that “CNP is widely expressed in multiple cell types in mouse and human kidneys including glomerular podocytes (results, first paragraph)”. This information cannot be found in the papers. There may be a misunderstanding because “CNP” is mentioned in the gene lists. However, this is the gene of a “cyclic nucleotide phoshodiesterase” and not CNP whose gene name is NPPC. In addition, the references are used as evidence for “for the formation of cGMP in podocytes by the NPRB” (results, first paragraph). This is also not correct. Please correct me if I am wrong.

The references 14 and 16 are single cell RNA sequencing studies. To obtain results for specific mRNAs investigators need to query the transcriptomics databases that accompany the manuscripts.

Database for these results, are available at the following websites: 1. http://humphreyslab.com/SingleCell/ and 2. https://susztaklab.com/EH/park/. I have also added these weblinks to each manuscript citing single cell RNA sequencing studies in the Reference Section. I am also requesting guidance how to reference these results in the manuscript.

4. It should be briefly discussed that the blood pressure values show a very high variability. Although this can occasionally occur with automated tail cuff measurement and is therefore not very unusual, it significantly weakens the authors' conclusion that there are no blood pressure differences between the genotypes.

The tail cuff blood pressure measurement did show variability in the results, which reduces chances of detecting a difference between groups. We can only speculate on the causes, but the major finding in the BP studies is the lack of a difference in the Gq mice compared to the Gq-KO mice. This is important because a difference in BP could affect the severity of the kidney disease. The comparison in these 2 groups (GQ and Gq-KO) found very similar BP levels suggesting that a difference in BP did not affect the in vivo findings in the study.

5. Legend Figure 2B: “KO mice” should be corrected to “KD cells”

We have corrected this mistake in the text of the manuscript.

Additional issues: PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

I do not think is necessary to publish the full review process.

References

1. Kubiak-Wlekly A, Perkowska-Ptasinska A, Olejniczak P, Rochowiak A, Kaczmarek E, Durlik M, et al. The Comparison of the Podocyte Expression of Synaptopodin, CR1 and Neprilysin in Human Glomerulonephritis: Could the Expression of CR1 be Clinically Relevant? Int J Biomed Sci. 2009;5(1):28-36. PubMed PMID: 23675111; PubMed Central PMCID: PMCPMC3614758.

2. Debiec H, Guigonis V, Mougenot B, Decobert F, Haymann JP, Bensman A, et al. Antenatal membranous glomerulonephritis due to anti-neutral endopeptidase antibodies. N Engl J Med. 2002;346(26):2053-60. Epub 2002/06/28. doi: 10.1056/NEJMoa012895. PubMed PMID: 12087141.

Attachment

Submitted filename: Response-to-Reviewers.docx

pone.0319424.s004.docx (23KB, docx)

Decision Letter 3

Michael Bader

28 Jan 2025

PONE-D-24-36618R3Podocyte specific knockout of the natriuretic peptide clearance receptor is podocyte protective in focal segmental glomerulosclerosisPLOS ONE

Dear Dr. Spurney,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands.Reviewer 1 and myself had no problem to find and search the single-cell RNAseq databases you cited and we could find the gene CNP to be expressed, but it codes for cyclic nucleotide phoshodiesterase, not for C-type natriuretic peptide. However we did not find NPPC (the gene encoding C-type natriuretic peptide) to be mentioned as gene being expressed in kidney. Therefore, we invite you to submit a revised version of the manuscript that addresses this point by either making sure that these databases contain the information on NPPC expression or citing other supporting references for the renal expression of this gene encoding CNP to support your sentence “CNP is widely expressed in multiple cell types in mouse and human kidneys including glomerular podocytes".

Please submit your revised manuscript by Mar 14 2025 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org . When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols . Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols .

We look forward to receiving your revised manuscript.

Kind regards,

Michael Bader

Academic Editor

PLOS ONE

Journal Requirements:

Please review your reference list to ensure that it is complete and correct. If you have cited papers that have been retracted, please include the rationale for doing so in the manuscript text, or remove these references and replace them with relevant current references. Any changes to the reference list should be mentioned in the rebuttal letter that accompanies your revised manuscript. If you need to cite a retracted article, indicate the article’s retracted status in the References list and also include a citation and full reference for the retraction notice.

[Note: HTML markup is below. Please do not edit.]

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/ . PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org . Please note that Supporting Information files do not need this step.

PLoS One. 2025 Mar 10;20(3):e0319424. doi: 10.1371/journal.pone.0319424.r009

Author response to Decision Letter 3


30 Jan 2025

Jnauary30, 2025

To whom it may concern:

I am submitting the revised manuscript entitled "Podocyte specific knockout of the natriuretic peptide clearance receptor ameliorates focal segmental glomerulosclerosis” for consideration for publication in Plos One. I am responding to the following comment:

“Reviewer 1 and myself had no problem to find and search the single-cell RNAseq databases you cited, and we could find the gene CNP to be expressed, but it codes for cyclic nucleotide phosphodiesterase, not for C-type natriuretic peptide. However, we did not find NPPC (the gene encoding C-type natriuretic peptide) to be mentioned as gene being expressed in kidney.”

We have retained the citations in the manuscript relevant to CNP expression in the kidney, but we have removed references to CNP expression in then following data bases:

- Reverence 14: https://susztaklab.com/EH/park/

- Reference 16: http://humphreyslab.com/SingleCell/

- Reference 50: http://humphreyslab.com/SingleCell/

These data bases are relevant to expression of natriuretic peptide receptors (NPRA, NPRB and NPRC) and these references remain in the manuscript citations. Changes to text of the manuscript have been highlighted in red.

Thanks for the careful review.

Correspondence regarding the manuscript should be sent to the following address:

Robert F. Spurney, M.D.

MSRB 2, Room 2013

106 Research Drive

Durham, N.C. 27710

(919)-684-9729

FAX: (919)-684-3011

E-mail: robert.spurney@duke.edu

Attachment

Submitted filename: Response-to-Reviewers_auresp_4.docx

pone.0319424.s005.docx (23KB, docx)

Decision Letter 4

Michael Bader

2 Feb 2025

Podocyte specific knockout of the natriuretic peptide clearance receptor is podocyte protective in focal segmental glomerulosclerosis

PONE-D-24-36618R4

Dear Dr. Spurney,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice will be generated when your article is formally accepted. Please note, if your institution has a publishing partnership with PLOS and your article meets the relevant criteria, all or part of your publication costs will be covered. Please make sure your user information is up-to-date by logging into Editorial Manager at Editorial Manager®  and clicking the ‘Update My Information' link at the top of the page. If you have any questions relating to publication charges, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Michael Bader

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Acceptance letter

Michael Bader

PONE-D-24-36618R4

PLOS ONE

Dear Dr. Spurney,

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now being handed over to our production team.

At this stage, our production department will prepare your paper for publication. This includes ensuring the following:

* All references, tables, and figures are properly cited

* All relevant supporting information is included in the manuscript submission,

* There are no issues that prevent the paper from being properly typeset

If revisions are needed, the production department will contact you directly to resolve them. If no revisions are needed, you will receive an email when the publication date has been set. At this time, we do not offer pre-publication proofs to authors during production of the accepted work. Please keep in mind that we are working through a large volume of accepted articles, so please give us a few weeks to review your paper and let you know the next and final steps.

Lastly, if your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

If we can help with anything else, please email us at customercare@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Prof. Michael Bader

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Data

    (PDF)

    pone.0319424.s001.pdf (1.4MB, pdf)
    Attachment

    Submitted filename: Respomse-to-Reviewers.pdf

    pone.0319424.s002.pdf (107.4KB, pdf)
    Attachment

    Submitted filename: PLOS Rebuttal Letter.docx

    pone.0319424.s003.docx (21.4KB, docx)
    Attachment

    Submitted filename: Response-to-Reviewers.docx

    pone.0319424.s004.docx (23KB, docx)
    Attachment

    Submitted filename: Response-to-Reviewers_auresp_4.docx

    pone.0319424.s005.docx (23KB, docx)

    Data Availability Statement

    All relevant data are within the manuscript and its Supporting Information files.


    Articles from PLOS One are provided here courtesy of PLOS

    RESOURCES