Diabetic nephropathy (DN) is the leading cause of end-stage renal disease, and effective treatment modalities that fully address its molecular etiology are lacking. Prior studies support that the stress response protein REDD1 (regulated in development and DNA damage 1) contributes to the development of diabetes complications. This study investigated a potential role for REDD1 expression in podocytes in diabetes-induced podocyte loss and compromised glomerular filtration. Podocyte-specific REDD1 deletion protected against renal injury, as evidenced by reduced albuminuria, glomerular hypertrophy, and mesangial matrix deposition in streptozotocin (STZ)-induced diabetic mice. Podocyte-specific REDD1 expression was required for diabetes-induced reduction in slit diaphragm (SD) proteins podocin and nephrin. Notably, podocyte-specific REDD1 deletion protected against podocytopenia and preserved glomerular basement membrane and foot process architecture in diabetic mice. In the kidneys of diabetic mice and in human podocyte cultures exposed to hyperglycemic conditions, REDD1 was necessary for increased expression of the transient receptor potential canonical 6 (TRPC6) channel. More specifically, REDD1 promoted nuclear factor-κB–dependent transcription of TRPC6, intracellular calcium entry, and cytoskeletal remodeling under hyperglycemic conditions. Overall, the findings provide new insight into the role of podocyte-specific REDD1 expression in renal pathology and support the possibility that therapeutics targeting REDD1 in podocytes could be beneficial for DN.

Article Highlights

  • Diabetes-induced albuminuria and reduced glomerular slit diaphragm proteins were associated with increased kidney REDD1 protein abundance.

  • Podocyte-specific deletion of REDD1 attenuated diabetes-induced slit diaphragm protein reduction and podocyte loss.

  • REDD1 was required for nuclear factor-κB–dependent TRPC6 expression and increased cytoplasmic calcium levels in podocytes.

  • Podocyte-specific expression of REDD1 was necessary for altered glomerular architecture and albuminuria in diabetic mice.

Podocyte injury and death are major contributing factors in the pathogenesis of diabetic nephropathy (DN). Podocyte loss is a reliable early indicator of renal injury, as it is observed in patients with early and late stages of DN (1,2) and in animal models of type 1 and type 2 diabetes (3,4). Podocytes are terminally differentiated epithelial cells with specialized cell junctions known as the slit diaphragm (SD). The SD connects interdigitating podocyte foot processes and establishes a size-selective barrier to protein loss (5). The SD is a zipper-like structure made by molecular cross-linking via proteins that include podocin and nephrin. The SD not only serves as molecular sieve but also acts as a complex signaling platform that regulates podocyte function (5,6). In diseased states like diabetes, the expression of SD proteins is reduced, resulting in compromised barrier function and albuminuria (7,8). Thus, an improved understanding of the specific molecular events that cause changes in the SD may identify new therapeutic targets for addressing DN pathogenesis.

Another key component of the SD that does not participate directly in cell junction is the cation channel transient receptor potential canonical channel 6 (TRPC6). TRPC6 is essential for maintaining calcium homeostasis in podocytes (9). TRPC6 activation enhances the influx of calcium ions in podocytes to influence cellular processes, including cytoskeletal organization and apoptosis (10). TRPC6 activation is linked to hereditary and acquired forms of proteinuric kidney diseases (11). Indeed, TRPC6 expression and activity are upregulated in DN (12); however, the specific signaling events responsible for diabetes-induced TRPC6 upregulation remain to be identified. One possibility is that activation of the transcription factor nuclear factor-κB (NF-κB) in response to diabetes influences TRPC6 in podocytes, given that prior reports provide evidence that NF-κB acts as a regulatory effector upstream of TRPC6 in other contexts (13,14).

The stress response protein regulated in development and DNA damage response 1 (REDD1; also known as DDIT4 or RTP801) promotes NF-κB signaling in response to diabetes (15). REDD1 has been linked to the development of diabetes complications, including DN (3,16). REDD1 protein abundance is increased in the kidney of patients with diabetes (16), in the kidney of diabetic mice, and in podocyte cultures exposed to hyperglycemic conditions (3,17). Remarkably, whole-body REDD1 deletion prevents podocyte loss and albuminuria in diabetic mice (3). In response to hyperglycemic conditions, REDD1 contributes to oxidative stress and proinflammatory signaling in podocytes that are potentially responsible for cell death (3,17,18), raising the question of whether diabetes-induced REDD1 expression specifically in podocytes is sufficient to drive renal pathology. Herein, we investigated the role of podocyte-specific REDD1 in DN.

Please see the Supplementary Material for detailed methods.

Animals

All procedures adhered to the National Institutes of Health Guide for the Care and Use of Laboratory Animals and were approved by the Penn State College of Medicine Institutional Animal Care and Use Committee. In male B6;129 mice that were homozygous for the REDD1 mutation (REDD1−/−) (19) or in wild-type (REDD1+/+) mice, diabetes was induced by administering 50 mg/kg streptozotocin (STZ) intraperitoneally for 5 consecutive days. Sodium citrate solution was administered as vehicle control. Diabetic phenotype was confirmed by fasting blood glucose concentrations >250 mg/dL. Male mice were used for all studies due to the resistance of female mice to developing hyperglycemia in response to low-dose STZ. To generate podocyte-specific REDD1 knockout (podKO) mice, floxed REDD1 mice (REDD1fl/fl) (20) were crossed with mice expressing Cre recombinase in podocytes [B6.Cg-Tg(NPHS2-cre)295Lbh/J; The Jackson Laboratory, stock no. 008205]. At 16 weeks of diabetes, mice were euthanized, and kidneys were processed as described below. Protein excretion and urine albumin-to-creatinine concentrations were determined from spot urine samples, as previously described (3).

Histology Studies

Formalin-fixed paraffin-embedded renal sections were processed for periodic acid Schiff (PAS) and hematoxylin and eosin (H&E) staining. Immunofluorescence labeling was done using the antibodies listed in Supplementary Table 1. Tissue sections were counterstained with 1 µmol/L Hoechst_33342 (Invitrogen), and photomicrographs were captured using a confocal laser microscope.

For transmission electron microscopy (TEM), kidneys were processed as described previously (3), and glutaraldehyde-fixed kidney cortical sections (65 nm) were mounted on a copper grid and photographed using a JEOL JEM1400 Transmission Electron Microscope located at the Penn State College of Medicine TEM Core Facility (RRID no.: SCR_021200).

Histomorphometry

All quantifications were performed in a masked manner. Mesangial matrix deposition and glomerular volume were evaluated as previously described (21). Glomerular tuft area (GA) was quantified using ImageJ and converted to glomerular volume (GV) by applying a spherical approximation formula (GV = 1.2545[GA]1.5). Mesangial index was defined as the ratio of mesangial area to glomerular tuft area.

ImageJ was used to examine electron photomicrographs. Quantification of glomerular basement membrane (GBM) thickness was performed using a modified method as previously described (22). GBM thickness was assessed by measuring 10 random and unbiased points in each image. To quantify foot process effacement, a method modified from van den Berg et al. (23) was used. The following formula was used to determine the mean width of the foot processes from each image:

Glomerular Isolation

Mouse glomeruli were isolated from kidney cortex using a differential adhesion method, as previously described (24). Briefly, the kidney cortex was digested with collagenase IV (1 mg/mL) in Hanks' balanced salt solution at 37°C for 15 min. Digested tissue was filtered successively, and differential adhesion of tubules was performed. The filtrate was washed on a 40-μm cell strainer, and intact glomeruli were isolated.

Cell Culture

Conditionally immortalized human podocytes (CIHP-1) were obtained from Dr. Moin Saleem (University of Bristol, Bristol, U.K.) and cultured as described (3). A stable CIHP-1 cell line deficient in REDD1 (REDD1 KO) was generated by clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) genome editing. To model hyperglycemia, cells were exposed to culture medium containing 30 mmol/L glucose (HG) or an osmotic control (OC; 11.1 mmol/L glucose plus 18.9 mmol/L mannitol). Cells were transfected using Lipofectamine 2000 (Life Technologies). Plasmids included pCMV5 vector, hemagglutinin (HA)-tagged pCMV-HA-REDD1, and pBabe-GFP-IKBalpha-mut (super repressor), which was a gift from William Hahn (Addgene plasmid no. 15264).

F-Actin Staining

F-actin fibers in CIHP-1 cells were stained with rhodamine phalloidin (Actistain 535, Cytoskeleton), nuclei were counterstained with 1 µmol/L DAPI (Invitrogen), and images were captured using a Leica DMi8 microscope. Ten fields containing at least 200 cells were randomly evaluated by two independent investigators, and each podocyte was rated on a 3-point scale, as described previously (25). The mean cortical F-actin score was then computed.

Cytosolic Calcium Imaging

Intracellular calcium (Ca2+) entry into the cytosol was measured by ratiometric imaging using fura-2, as described previously (26). In brief, podocytes were loaded with 2 µmol/L Fura-2/AM (no. F1221, Invitrogen), and fluorescence was measured using a Leica DMI 8 fluorescence microscope. Consecutive excitation at 340 nm (F340) and 380 nm (F380) was applied every 2 s, and emission fluorescence was collected at 505 nm for 3 min. To determine TRPC6-mediated Ca2+ entry, cells were treated with the TRPC6 selective agonist oleoyl-2-acetyl-sn-glycerol (OAG, 300 µmol/L; Cayman Chemicals). Intracellular Ca2+ flux was measured as F340-to-F380 ratios obtained from groups of >25 single cells from three independent experiments.

Protein Analysis

Total proteins were extracted from cells, renal cortical tissue, or isolated glomeruli using radioimmunoprecipitation assay lysis buffer. Western blot analysis was performed as previously described (3) with the appropriate antibodies (Supplementary Table 1).

PCR Analysis

Total RNA was extracted, reverse transcribed, and analyzed by real-time quantitative PCR (qPCR) (QuantStudio 12 K Flex Real-Time PCR System, RRID:SCR_021098) with primers listed in Supplementary Table 2. Mean cycle threshold values were determined. Fold change in mRNA expression relative to GAPDH mRNA was calculated.

Chromatin Immunoprecipitations

The binding capacity of NF-κB p65 to the promoter region of the TRPC6 gene was confirmed using a chromatin-immunoprecipitation (ChIP) assay in conjunction with qPCR (ChIP-qPCR). The ChIP assay was performed using a Simple ChIP Plus Enzymatic ChIP Kit (Cell Signaling Technology), following the manufacturer’s instructions. In brief, proteins from CIHP-1 podocytes were cross-linked with chromatin, and subsequently, the protein-chromatin complex was broken using ultrasonication. The soluble chromatin was incubated with anti-p65 antibody or IgG (negative control) at 4°C overnight, and immunoprecipitation was done using protein G magnetic beads (Cell Signaling Technology). The recovered DNA was then analyzed by qPCR using primers listed in Supplementary Table 2. Results were tabulated as fold enrichment normalized to the IgG controls (34).

Statistical Analysis

Data are expressed as mean ± SD. Statistical analysis of data with more than two groups was performed by two-way ANOVA, with the Tukey test for multiple comparisons used for pairwise analysis. The relationship between the urine albumin-to-creatinine ratio (ACR) and REDD1 protein abundance was tested by Spearman correlation analysis. Significance was defined as P < 0.05 for all analyses. Sample sizes for each experiment are listed in Supplementary Table 3, and exact P values for significantly different groups are listed in Supplementary Table 4.

Data and Resource Availability

Primary data, including unedited Western blot images and immunofluorescent images supporting the findings of this study, are reported within the supplementary information. Animation files for calcium imaging are available at https://doi.org/10.6084/m9.figshare.26790010.v1. Excel files with raw data supporting the findings will be made available upon request.

REDD1 Expression Is Required for Renal Injury in DN

In the kidney of STZ-diabetic mice, REDD1 protein abundance was increased in coordination with elevated urinary albumin (Fig. 1A) (3). In support of prior reports (7), reduced Nphs2 (podocin) mRNA expression (Fig. 1B) and podocin protein abundance (Fig. 1C and D) were observed in the kidneys of STZ-diabetic wild-type mice. REDD1 deletion reduced the diabetes-induced attenuation in podocin mRNA and protein in the kidney. A similar REDD1-dependent reduction in NPHS2 mRNA expression (Fig. 1E) and podocin protein abundance (Fig. 1F and G) was also seen in human podocytes exposed to hyperglycemic culture conditions. To confirm the impact of REDD1 on podocin abundance in podocytes, REDD1 was restored in REDD1-deficient cells by expression of the HA-tagged REDD1 plasmid. Ectopic REDD1 expression reduced podocin levels in REDD1 KO cells when exposed to OC or hyperglycemic conditions (Fig. 1H). Thus, increased REDD1 protein content was sufficient to attenuate podocin protein abundance in podocytes.

Figure 1

REDD1 promoted diabetes-induced renal injury. A–D: Wild-type (WT; REDD1+/+) and REDD1 KO (REDD1−/−) mice were administered STZ to induce diabetes. Nondiabetic mice received vehicle (Veh) control. Analysis was performed after 16 weeks of diabetes. A: REDD1 protein abundance in kidney homogenates was positively correlated with urine ACR in WT mice administered Veh or STZ. REDD1 protein and urine ACR were evaluated by Western blotting and ELISA, respectively (3). B: Nphs2 mRNA expression in renal cortical homogenates was determined by qPCR. Podocin protein was evaluated in renal sections by immunofluorescence (C) and Western blotting of renal cortical homogenates (D). E–H: Differentiated WT or REDD1 CRISPR KO CIHP-1 cells were exposed to culture medium containing 30 mmol/L glucose (HG) or 5 mmol/L glucose plus 25 mmol/L mannitol as an OC for 48 h. E: NPHS2 mRNA expression in CIHP-1 cells was quantified by qPCR. F: Cells were immunolabeled for podocin. Podocin localization in the cytosol is indicated in zoomed inset (white arrowheads). G: Podocin protein abundance was quantified by Western blotting. H: Podocin protein abundance was evaluated in REDD1 KO CIHP-1 cells expressing an empty vector (EV) control or HA-tagged REDD1. Representative photomicrographs (scale bar: 50 µm) and Western blots with molecular weights in kDa to the right of the blot are shown. Protein abundance is expressed relative to actin or GAPDH and is normalized to the amount observed in nondiabetic WT control mice or in cells exposed to an OC. Individual data points (n = 4–6) are graphed and presented as means ± SD. Statistical significance was determined by two-way ANOVA with Tukey post hoc analysis. *P < 0.05 vs. Veh or OC; #P < 0.05 vs. REDD1+/+, WT, or EV.

Figure 1

REDD1 promoted diabetes-induced renal injury. A–D: Wild-type (WT; REDD1+/+) and REDD1 KO (REDD1−/−) mice were administered STZ to induce diabetes. Nondiabetic mice received vehicle (Veh) control. Analysis was performed after 16 weeks of diabetes. A: REDD1 protein abundance in kidney homogenates was positively correlated with urine ACR in WT mice administered Veh or STZ. REDD1 protein and urine ACR were evaluated by Western blotting and ELISA, respectively (3). B: Nphs2 mRNA expression in renal cortical homogenates was determined by qPCR. Podocin protein was evaluated in renal sections by immunofluorescence (C) and Western blotting of renal cortical homogenates (D). E–H: Differentiated WT or REDD1 CRISPR KO CIHP-1 cells were exposed to culture medium containing 30 mmol/L glucose (HG) or 5 mmol/L glucose plus 25 mmol/L mannitol as an OC for 48 h. E: NPHS2 mRNA expression in CIHP-1 cells was quantified by qPCR. F: Cells were immunolabeled for podocin. Podocin localization in the cytosol is indicated in zoomed inset (white arrowheads). G: Podocin protein abundance was quantified by Western blotting. H: Podocin protein abundance was evaluated in REDD1 KO CIHP-1 cells expressing an empty vector (EV) control or HA-tagged REDD1. Representative photomicrographs (scale bar: 50 µm) and Western blots with molecular weights in kDa to the right of the blot are shown. Protein abundance is expressed relative to actin or GAPDH and is normalized to the amount observed in nondiabetic WT control mice or in cells exposed to an OC. Individual data points (n = 4–6) are graphed and presented as means ± SD. Statistical significance was determined by two-way ANOVA with Tukey post hoc analysis. *P < 0.05 vs. Veh or OC; #P < 0.05 vs. REDD1+/+, WT, or EV.

Close modal

Podocyte Expression of REDD1 Was Required for Glomerular Injury in DN

Hemizygous podocin-cre mice were crossed with REDD1fl/fl mice to achieve podocyte-specific recombination (NPHS2-cre+, REDD1fl/fl; REDD1 podKO). PCR analysis of genomic DNA was used to confirm homozygous expression of the floxed REDD1 gene and NPHS2-Cre in REDD1 podKO mice (Supplementary Fig. 1A). Compared with REDD1fl/fl mice, podocyte-specific REDD1 deletion nearly eliminated REDD1 mRNA expression in isolated glomeruli and partially reduced REDD1 mRNA expression in kidney homogenates from REDD1 podKO mice (Fig. 2A). REDD1 mRNA expression in liver, heart, and retina were unchanged between the two groups. To evaluate the role of podocyte-specific REDD1 expression on diabetes-induced renal injury, mice were administered STZ. Diabetic and nondiabetic REDD1 podKO mice exhibited similar fasting blood glucose concentrations as those observed in diabetic and nondiabetic REDD1fl/fl mice, respectively (Fig. 2B). REDD1 protein abundance was increased within glomeruli of diabetic REDD1fl/fl mice compared with nondiabetic controls (Fig. 2C and D). Podocyte-specific REDD1 deletion reduced glomerular REDD1 protein abundance and prevented an increase in REDD1 in response to diabetes. Diabetes reduced Nphs2 mRNA expression within glomeruli isolated from REDD1fl/fl mice, and the suppressive effect was blunted in REDD1 podKO mice (Fig. 2E). Similarly, glomerular podocin protein levels were also reduced in diabetic REDD1fl/fl mice but not in diabetic REDD1 podKO mice compared with their respective nondiabetic controls (Fig. 2F and G). The data support that podocyte-specific REDD1 contributes to diabetes-induced reduction in podocin.

Figure 2

Podocyte-specific deletion of REDD1 prevented podocyte injury in diabetic mice. A: REDD1 mRNA expression was quantified in glomerular isolates (GL) and in kidney, heart, liver, and retina by qPCR. B–G: REDD1fl/fl and REDD1 podKO mice were administered STZ or a vehicle (Veh) control. Analysis was performed after 16 weeks of diabetes. B: Fasting blood glucose (FBG) levels were measured. REDD1 (C) and podocin (F) were visualized in renal sections by immunofluorescence microscopy (scale bar: 50 µm). Nuclei were counterstained with Hoechst_33342. Protein abundance of REDD1 (D) and podocin (G) was quantified in glomerular isolates by Western blotting. Nphs2 mRNA expression was analyzed in glomerular isolates by qPCR (E). Representative blots are shown with molecular weights in kDa indicated to the right of the blot. Protein abundance is expressed relative to actin and is normalized to the amount observed in nondiabetic REDD1fl/fl control mice. Data are presented as means ± SD, and individual data points are plotted (n = 3–6). Statistical significance was determined by two-way ANOVA with Tukey post hoc analysis. *P < 0.05 vs. Veh; #P < 0.05 vs. REDD1fl/fl.

Figure 2

Podocyte-specific deletion of REDD1 prevented podocyte injury in diabetic mice. A: REDD1 mRNA expression was quantified in glomerular isolates (GL) and in kidney, heart, liver, and retina by qPCR. B–G: REDD1fl/fl and REDD1 podKO mice were administered STZ or a vehicle (Veh) control. Analysis was performed after 16 weeks of diabetes. B: Fasting blood glucose (FBG) levels were measured. REDD1 (C) and podocin (F) were visualized in renal sections by immunofluorescence microscopy (scale bar: 50 µm). Nuclei were counterstained with Hoechst_33342. Protein abundance of REDD1 (D) and podocin (G) was quantified in glomerular isolates by Western blotting. Nphs2 mRNA expression was analyzed in glomerular isolates by qPCR (E). Representative blots are shown with molecular weights in kDa indicated to the right of the blot. Protein abundance is expressed relative to actin and is normalized to the amount observed in nondiabetic REDD1fl/fl control mice. Data are presented as means ± SD, and individual data points are plotted (n = 3–6). Statistical significance was determined by two-way ANOVA with Tukey post hoc analysis. *P < 0.05 vs. Veh; #P < 0.05 vs. REDD1fl/fl.

Close modal

REDD1 Deletion in Podocytes Prevented Diabetes-Induced Glomerular Pathology

Compared with nondiabetic mice, diabetic mice had reduced body weights (Supplementary Fig. 1B) but increased kidney-to-body weight ratio (Supplementary Fig. 1C). No genotype differences were observed. Glomerular morphology and mesangial proliferation were examined by H&E and PAS staining, respectively (Fig. 3A). Histology revealed increased glomerular size, basement membrane thickening, and mesangial matrix deposition in diabetic REDD1fl/fl mice compared with nondiabetic controls. Podocyte deletion of REDD1 prevented increased glomerular volume and reduced the Bowman space in the kidneys of diabetic mice (Fig. 3B). Increased mesangial proliferation was also attenuated in mice lacking podocyte REDD1 postdiabetes induction (Fig. 3C). Podocytopenia is a clinical hallmark of early DN in patients with type 1 or type 2 diabetes (1,2). Wilms tumor-1 (WT-1) immunolabeling was used to identify podocytes. Podocyte-specific ablation of REDD1 attenuated diabetes-induced podocyte loss and prevented reduced nephrin protein abundance compared with diabetic REDD1fl/fl mice (Fig. 3D). Quantification of WT-1–positive nuclei within glomeruli revealed a reduction in podocytes within glomeruli of diabetic REDD1fl/fl mice but not in diabetic REDD1 podKO mice (Fig. 3E). Podocyte apoptosis was also characterized by visualizing TUNEL-positive nuclei colocalized with WT-1 within the glomerulus (Fig. 3F). Whereas an increase in TUNEL-positive cells colocalized with WT-1 cell staining was present in glomeruli of diabetic REDD1fl/fl mice, an increase in apoptotic cells was attenuated in diabetic REDD1 podKO mice (Fig. 3G). In summary, REDD1 deletion in podocytes was sufficient to prevent development of glomerular pathology and podocytopenia in DN.

Figure 3

Podocyte expression of REDD1 contributed to podocyte loss and the development of renal pathology. REDD1fl/fl and REDD1 podKO mice were administered STZ or vehicle (Veh) control, and kidneys were analyzed 16 weeks after diabetes induction. A: Renal pathology was characterized by H&E and PAS staining. Glomerular tuft volume (B) (n = 5; 20 per section) and mesangial proliferation (C) (n = 5; 15 per section) were quantified. D: Colocalization of podocyte marker WT-1 and nephrin was visualized by immunofluorescence. E: WT-1–positive cells were counted (n = 5; 20 per section). F: Colocalization of TUNEL-positive apoptotic nuclei with podocyte markers WT-1 and nephrin was evaluated by immunofluorescence. G: Cells positive for both TUNEL and WT-1 were counted (n = 3; 10 per section). Representative photomicrographs are shown (scale bar: 50 µm). Individual means of each n are plotted and graphed as means ± SD. Statistical significance was determined using two-way ANOVA and the Tukey post hoc test. *P < 0.05 vs. Veh; #P < 0.05 vs. REDD1fl/fl.

Figure 3

Podocyte expression of REDD1 contributed to podocyte loss and the development of renal pathology. REDD1fl/fl and REDD1 podKO mice were administered STZ or vehicle (Veh) control, and kidneys were analyzed 16 weeks after diabetes induction. A: Renal pathology was characterized by H&E and PAS staining. Glomerular tuft volume (B) (n = 5; 20 per section) and mesangial proliferation (C) (n = 5; 15 per section) were quantified. D: Colocalization of podocyte marker WT-1 and nephrin was visualized by immunofluorescence. E: WT-1–positive cells were counted (n = 5; 20 per section). F: Colocalization of TUNEL-positive apoptotic nuclei with podocyte markers WT-1 and nephrin was evaluated by immunofluorescence. G: Cells positive for both TUNEL and WT-1 were counted (n = 3; 10 per section). Representative photomicrographs are shown (scale bar: 50 µm). Individual means of each n are plotted and graphed as means ± SD. Statistical significance was determined using two-way ANOVA and the Tukey post hoc test. *P < 0.05 vs. Veh; #P < 0.05 vs. REDD1fl/fl.

Close modal

REDD1 Promoted TRPC6 Protein Abundance and Changes in Podocyte Structure in Response to Hyperglycemic Conditions

Foot processes function and the preservation of SD integrity depend on focal podocyte adhesions and actin cytoskeletal structure (27). To determine whether REDD1 contributed to impairment of podocyte cytoskeletal architecture, we assessed podocyte F-actin arrangement. Hyperglycemic conditions lead to overt F-actin disorganization in wild-type podocytes, which was characterized by the loss of dispersed central stress fibers and the appearance of cortical F-actin (Fig. 4A and B). Unlike wild-type cells, REDD1-deficient cells maintained their actin cytoskeletal structure after exposure to hyperglycemic environments. Calcium (Ca2+) homeostasis is important in regulating podocyte cytoskeleton dynamics, and hyperglycemic conditions increase cytosolic Ca2+ entry in podocytes (12). Upon exposure to hyperglycemic conditions, basal Ca2+ levels were increased in wild-type podocytes but not in REDD1-deficient podocytes (Fig. 4C and D). In wild-type cells exposed to hyperglycemic conditions, TRPC6 protein abundance was increased (Fig. 4E). However, a similar increase in TRPC6 was not observed in REDD1-deficinet cells. Ca2+ entry in response to the TRPC6-selective agonist OAG was augmented in wild-type cells exposed to hyperglycemic conditions (Fig. 4F). REDD1 deletion reduced OAG-mediated Ca2+ entry and prevented an increase in the response under hyperglycemic conditions. To determine whether REDD1 had a similar effect in the kidney, renal sections were immunolabeled for TRPC6. Diabetes increased TRPC6 colocalization with nephrin within glomeruli of REDD1fl/fl mice but not in REDD1 podKO mice (Fig. 4G). Western blot analysis of glomerular protein isolates from diabetic mice also showed a REDD1-dependent increase in TRPC6 protein compared with nondiabetic controls (Fig. 4H).

Figure 4

REDD1 promoted cytoskeletal remodeling and increased TRPC6 protein abundance in podocytes exposed to hyperglycemic conditions. A–F: Differentiated wild-type (WT) and REDD1 KO CIHP-1 cells were exposed to media containing 30 mmol/L glucose (HG) or 5 mmol/L glucose plus 25 mmol/L mannitol OC for 48 h. A: Actin assembly was visualized by staining cells with rhodamine phalloidin (red). B: Cortical F-actin scores (CFS) were quantified (n = 3, 200 cells each). C: Cytosolic Ca2+ levels were measured by imaging using fura-2 AM (the ratio of cells with the graphed response to the TRPC6 agonist OAG is indicated for each condition). D: Baseline Ca2+ levels were compared in podocyte cultures. E: TRPC6 protein abundance was quantified in cell lysates by Western blotting. F: Peak Ca2+ entry responses after the addition of OAG (300 µmol/L) were plotted. G: Renal sections from STZ-diabetic or nondiabetic (Veh) REDD1fl/fl and REDD1 podKO mice were immunolabeled for TRPC6 and nephrin. H: TRPC6 protein was quantified in glomerular isolates by Western blotting. Representative photomicrographs (scale bar: 50 µm; inset in G: 25 µm) and Western blots are shown with protein mass in kDa indicated to the right of each blot. Protein abundance is expressed relative to GAPDH or actin and is normalized to the amount observed in WT cells exposed to an OC or nondiabetic REDD1fl/fl control mice. Data in A, B, C, E, and H are graphed as means ± SD. Data in D and F are represented as box and whisker plots. Statistical significance was determined using two-way ANOVA and the Tukey post hoc test. *P < 0.05 vs. OC or Veh; #P < 0.05 vs. WT or REDD1fl/fl.

Figure 4

REDD1 promoted cytoskeletal remodeling and increased TRPC6 protein abundance in podocytes exposed to hyperglycemic conditions. A–F: Differentiated wild-type (WT) and REDD1 KO CIHP-1 cells were exposed to media containing 30 mmol/L glucose (HG) or 5 mmol/L glucose plus 25 mmol/L mannitol OC for 48 h. A: Actin assembly was visualized by staining cells with rhodamine phalloidin (red). B: Cortical F-actin scores (CFS) were quantified (n = 3, 200 cells each). C: Cytosolic Ca2+ levels were measured by imaging using fura-2 AM (the ratio of cells with the graphed response to the TRPC6 agonist OAG is indicated for each condition). D: Baseline Ca2+ levels were compared in podocyte cultures. E: TRPC6 protein abundance was quantified in cell lysates by Western blotting. F: Peak Ca2+ entry responses after the addition of OAG (300 µmol/L) were plotted. G: Renal sections from STZ-diabetic or nondiabetic (Veh) REDD1fl/fl and REDD1 podKO mice were immunolabeled for TRPC6 and nephrin. H: TRPC6 protein was quantified in glomerular isolates by Western blotting. Representative photomicrographs (scale bar: 50 µm; inset in G: 25 µm) and Western blots are shown with protein mass in kDa indicated to the right of each blot. Protein abundance is expressed relative to GAPDH or actin and is normalized to the amount observed in WT cells exposed to an OC or nondiabetic REDD1fl/fl control mice. Data in A, B, C, E, and H are graphed as means ± SD. Data in D and F are represented as box and whisker plots. Statistical significance was determined using two-way ANOVA and the Tukey post hoc test. *P < 0.05 vs. OC or Veh; #P < 0.05 vs. WT or REDD1fl/fl.

Close modal

REDD1 Is Required for Increased TRPC6 Transcription Under Hyperglycemic Conditions

Trpc6 mRNA expression in glomerular isolates from REDD1fl/fl mice was significantly increased with diabetes, and podocyte-specific deletion of REDD1 prevented the effect (Fig. 5A). A similar REDD1-dependent increase was also observed in podocyte cultures exposed to hyperglycemic conditions (Fig. 5B). NF-κB promotes transcription of TRPC6 by increased binding of the p65 subunit at the TRPC6 promoter (14). To determine the impact of NF-κB activity on TRPC6 transcription, wild-type cells expressing an empty vector or an I-κB superrepressor mutant were exposed to hyperglycemic conditions. Suppression of NF-κB activity prevented an increase in TRPC6 mRNA expression upon exposure to hyperglycemic conditions (Fig. 5C). To assess the impact of REDD1 on p65 NF-κB binding to the TRPC6 promoter, a ChIP PCR assay was performed. Increased binding of p65 at the TRPC6 promoter was observed in wild-type cells exposed to hyperglycemic conditions compared with OCs (Fig. 5D). REDD1 deletion suppressed p65 binding to the TRPC6 promoter and prevented an increase in TRPC6 enrichment in the p65-ChIP in cells exposed to hyperglycemic conditions.

Figure 5

REDD1 promoted NF-κB–dependent TRPC6 transcription. A: Trpc6 mRNA expression in the kidney of STZ-diabetic or nondiabetic (Veh) REDD1fl/fl and REDD1 podKO mice was quantified by qPCR 16 weeks after diabetes induction. B–D: Differentiated wild-type (WT) or REDD1 KO CIHP-1 cells were exposed to media containing 30 mmol/L glucose (HG) or 5 mmol/L glucose plus 25 mmol/L mannitol as an OC for 48 h. B: TRPC6 mRNA expression was evaluated by qPCR. C: TRPC6 mRNA expression was determined in CIHP-1 WT cells expressing an empty vector (EV) or I-κB super repressor (I-κB SR) plasmid. D: ChIP-PCR analysis was done in WT and REDD1 KO podocytes to quantify binding of p65 NF-κB in the promoter region of the TRPC6 gene. The p65 consensus motif on the TRPC6 promoter region is indicated in red. Individual data points are plotted and presented as means ± SD (n = 3–6). Differences between groups were identified by two-way ANOVA with Tukey post hoc analysis. *P < 0.05 vs. Veh or OC; #P < 0.05 vs. REDD1fl/fl, WT, or EV.

Figure 5

REDD1 promoted NF-κB–dependent TRPC6 transcription. A: Trpc6 mRNA expression in the kidney of STZ-diabetic or nondiabetic (Veh) REDD1fl/fl and REDD1 podKO mice was quantified by qPCR 16 weeks after diabetes induction. B–D: Differentiated wild-type (WT) or REDD1 KO CIHP-1 cells were exposed to media containing 30 mmol/L glucose (HG) or 5 mmol/L glucose plus 25 mmol/L mannitol as an OC for 48 h. B: TRPC6 mRNA expression was evaluated by qPCR. C: TRPC6 mRNA expression was determined in CIHP-1 WT cells expressing an empty vector (EV) or I-κB super repressor (I-κB SR) plasmid. D: ChIP-PCR analysis was done in WT and REDD1 KO podocytes to quantify binding of p65 NF-κB in the promoter region of the TRPC6 gene. The p65 consensus motif on the TRPC6 promoter region is indicated in red. Individual data points are plotted and presented as means ± SD (n = 3–6). Differences between groups were identified by two-way ANOVA with Tukey post hoc analysis. *P < 0.05 vs. Veh or OC; #P < 0.05 vs. REDD1fl/fl, WT, or EV.

Close modal

REDD1 Was Required for Deterioration of Podocyte Architecture and Decline in Filtration Function in Diabetic Mice

TEM was used to determine changes in glomerular ultrastructure. Compared with nondiabetic mice, we observed diffused thickening of the GBM and extensive fusion of foot processes within glomeruli of diabetic REDD1fl/fl mice (Fig. 6A). Morphometric analysis of TEM photomicrographs showed a significant increase in GBM thickness that was reduced in glomeruli of diabetic REDD1 podKO mice (Fig. 6B). We also observed increased podocyte foot processes width within the glomeruli of diabetic REDD1fl/fl mice compared with nondiabetic controls (Fig. 6C). Podocyte REDD1 deletion prevented foot process effacement in diabetic mice. Microalbuminuria is an important early indicator of DN progression and is often caused by the dysfunction of the glomerular filtration barrier (28). Urinalysis of diabetic REDD1fl/fl mice revealed an increased urinary protein band at 60 kDa that was reduced in the urine from diabetic REDD1 podKO mice (Fig. 6D). Quantification of urine albumin and creatinine levels showed a marked increase in urine ACR in diabetic REDD1fl/fl mice that was attenuated in diabetic REDD1 podKO mice (Fig. 6E). These data support that REDD1 deletion from podocytes is sufficient to attenuate the glomerular structural changes and filtration function decline that is observed in diabetic mice.

Figure 6

Podocyte-specific REDD1 deletion attenuated glomerular pathology and filtration function in diabetic mice. REDD1fl/fl and REDD1 podKO mice were administered STZ or a vehicle (Veh) control, and kidneys were analyzed 16 weeks after diabetes induction. A: Photomicrographs of TEM sections of podocytes are shown (scale bar: 1 µm). Box indicates the area shown at increased magnification below each image. Foot processes with SDs (arrowheads), foot process effacement (arrows), and GBM are indicated. Thickness of GBM (B) and foot process width (C) were calculated. D: Urinary proteins were separated by SDS-PAGE and visualized by protein staining. Protein mass in kDa is indicated at left. E: Urine ACR was determined. Individual data are plotted and presented as means ± SD. Significance was determined using two-way ANOVA and the Tukey post hoc test. *P < 0.05 vs. Veh; #P < 0.05 vs. REDD1fl/fl.

Figure 6

Podocyte-specific REDD1 deletion attenuated glomerular pathology and filtration function in diabetic mice. REDD1fl/fl and REDD1 podKO mice were administered STZ or a vehicle (Veh) control, and kidneys were analyzed 16 weeks after diabetes induction. A: Photomicrographs of TEM sections of podocytes are shown (scale bar: 1 µm). Box indicates the area shown at increased magnification below each image. Foot processes with SDs (arrowheads), foot process effacement (arrows), and GBM are indicated. Thickness of GBM (B) and foot process width (C) were calculated. D: Urinary proteins were separated by SDS-PAGE and visualized by protein staining. Protein mass in kDa is indicated at left. E: Urine ACR was determined. Individual data are plotted and presented as means ± SD. Significance was determined using two-way ANOVA and the Tukey post hoc test. *P < 0.05 vs. Veh; #P < 0.05 vs. REDD1fl/fl.

Close modal

Podocyte injury, podocytopenia, and, consequentially, microalbuminuria are hallmarks of early DN. Using a combination of in vivo and in vitro loss-of-function approaches, we demonstrated herein that podocyte-specific expression of the stress response protein REDD1 was required for podocyte loss, deterioration of the glomerular filtration barrier, and the subsequent decline in filtration function in a well-characterized preclinical model of type 1 diabetes. The studies here also provide evidence that REDD1 in podocytes acts to augment TRPC6-mediated intracellular Ca2+ influx, disorganization of the actin cytoskeleton, and reduced SD integrity. Overall, the findings support a model wherein diabetes-induced podocyte-specific REDD1 expression disrupts calcium homeostasis, resulting in glomerular injury and filtration function deficits (Fig. 7).

Figure 7

Podocyte-specific REDD1 contributes to diabetes-induced glomerular injury and filtration defects. Diabetes-enhanced podocyte REDD1 protein abundance leading to NF-κB–mediated upregulation of TRPC6 expression, increased cytosolic calcium entry, and altered cytoskeletal organization. Podocyte-specific REDD1 expression was necessary for compromised glomerular architecture and albuminuria.

Figure 7

Podocyte-specific REDD1 contributes to diabetes-induced glomerular injury and filtration defects. Diabetes-enhanced podocyte REDD1 protein abundance leading to NF-κB–mediated upregulation of TRPC6 expression, increased cytosolic calcium entry, and altered cytoskeletal organization. Podocyte-specific REDD1 expression was necessary for compromised glomerular architecture and albuminuria.

Close modal

A growing body of evidence suggests that REDD1 expression in the kidney contributes to the development of DN pathophysiology (3,16,17). Single-cell sequencing provides evidence that REDD1 expression is not restricted to a specific type of kidney cell (29). We previously reported that REDD1 expression was increased within the renal cortex of STZ-diabetic mice and that germline deletion of REDD1 prevented diabetes-induced podocyte loss (3). Moreover, REDD1 deletion in cultured podocytes prevented podocyte apoptosis in response to hyperglycemic conditions (3). Because podocyte loss is a clinical hallmark of early DN pathogenesis, we used Cre-lox recombination to KO REDD1 specifically in podocytes. REDD1 deletion in podocytes nearly eliminated REDD1 expression within glomerular isolates and attenuated REDD1 levels in renal cortical homogenates. In further support of podocyte specificity, REDD1 expression in other tissues, including the liver, retina, and heart, remained unchanged. Although the NPHS2-cre is a well-established tool to modify gene function precisely in podocytes (30), it has been reported that mice expressing the NPHS2-cre demonstrate modest GBM thickening and altered foot process morphology (31). We did not observe a change in these end points in nondiabetic REDD1 podKO mice compared with nondiabetic REDD1fl/fl mice. Rather, podocyte-specific deletion of REDD1 in mice expressing NPHS2-cre reduced the diabetes-induced changes in GBM and foot process morphology. Importantly, REDD1 deletion in podocytes was sufficient to prevent early renal pathology and attenuate podocyte loss with STZ-induced diabetes. Notably, single-nucleus and single-cell transcriptomic analyses of human kidneys have indicated REDD1 expression in other cell types, including endothelial cells and immune cell populations (32). However, REDD1 deletion in podocytes was sufficient to reduce REDD1 levels in both glomerular isolates and renal cortical homogenates from the kidneys of diabetic mice. This supports that REDD1-dependent processes in podocytes are potentially necessary for REDD1 upregulation in other renal cells in the context of diabetes.

Podocyte injury is a key event in the development of glomerular filtration defects. In the current study, we used a well-established model of type 1 diabetes to examine this phenomenon. A limitation of this study is that compared with other inbred mouse strains (e.g., DBA/2J, KK-Ay), C57BL/6 mice with STZ-induced hypoinsulinemia and secondary hyperglycemia have modest changes in glomerular pathology and mild-to-moderate increases in urine albumin excretion (33,34). Whereas DBA/2 mice develop albuminuria within 5 weeks of diabetes induction, the onset of albuminuria with STZ-diabetes is delayed in C57BL/6 mice (33). Importantly, low-dose STZ has no direct toxic effects on podocytes after 2 weeks of administration (35). Consistent with our prior report (3), we observed a close association between renal REDD1 levels and increased albumin excretion. Remarkably, podocyte-specific REDD1 deletion was sufficient to maintain the structural and functional integrity of the glomerular filtration barrier and prevented albuminuria in STZ-diabetic mice. Together, the observations support a key role of podocyte REDD1 expression in the failed adaptive response of the kidney to diabetes.

In diseased states like diabetes, expression of SD proteins is reduced, resulting in compromised barrier function and albuminuria. In support of prior observations (10), SD proteins, including podocin and nephrin, were significantly attenuated by diabetes or exposure to hyperglycemic conditions. We also observed that the reduction in SD protein content was concomitant with compromised glomerular ultrastructure. REDD1 deletion in podocytes preserved SD protein levels and prevented the deterioration of podocyte architecture in response to diabetogenic stimuli. Podocyte-specific REDD1 deletion also prevented characteristic signs of early DN pathology that included increased volume of the glomerular tuft, GBM thickening, foot process effacement, and podocyte apoptosis (7).

TRPC6 is a stress-induced cation channel that associates with the SD and is required for proper renal function (36). Clinical observations and gain-of-function mutation studies demonstrate that increased TRPC6 expression correlates with aberrant calcium homeostasis and proteinuric kidney disease (11). In metabolic diseases like diabetes, TRPC6 expression is associated with reduced SD protein expression and disorganized F-actin assembly that leads to foot process effacement, podocyte loss, and eventually, the development of proteinuria (10,37,38). Hyperglycemia has previously been implicated as a major driver of TRPC6 expression and activity (10,38). Herein, REDD1 was required for increased TRPC6 expression and an increase in cytoplasmic Ca2+ in podocytes exposed to hyperglycemic conditions, suggesting a potential mechanism by which REDD1 regulates podocyte dysfunction. It is worth noting that the data herein do not exclude the possibility that REDD1 also acts to influences TRPC6 localization or activity.

Although some reports suggest that NF-κB mediates suppression of canonical TRPC6 expression (13), others have shown that the p65 subunit of NF-κB binds to the promoter region of TRPC6 to increase its transcription (14). An abundance of literature in recent years has supported a role for REDD1 in mediating NF-κB–mediated inflammatory response in diabetes complications (39–43). In response to hyperglycemic conditions, NF-κB suppression prevented increased TRPC6 mRNA expression. Notably, ChIP PCR analysis indicated that REDD1 deletion in podocytes reduced NF-κB binding to the TRPC6 promoter and prevented an increase in response to hyperglycemic conditions. It is worth noting that other transcription factors, including AP-1 and SP-1, share consensus-binding motifs in the TRPC6 promoter region and might also influence the expression of the TRPC6 gene through NF-κB–independent mechanisms (13). Moreover, studies in multiple cell types, including podocytes (37,44–46) and vascular myocytes (47), attribute a role for increased oxidative stress in the activation and expression of the TRPC6 channel. However, studies have also shown that reactive oxygen species–dependent activation of protein kinase C in mesangial cells can reduce TRPC6 expression (13).

Overall, the data herein are consistent with a model wherein enhanced REDD1 expression in podocytes leads to their failure to properly adapt to the diabetic metabolic environment, resulting in podocytopenia and microalbuminuria. The results provide new insight into mechanisms of podocyte injury and support targeting REDD1 as a therapeutic approach in the context of DN. Inhibiting REDD1 could also have additional benefits on account of its known inhibitory action on GSK3β activity (3,40,48). Although targeting REDD1 using siRNA therapeutics has previously been used in a clinical setting to target retinal complications such as macular degeneration (49) and diabetic macular edema (50), the efficacy and safety of inhibiting REDD1 as a potential strategy for DN remains to be established.

See accompanying article, p. 265.

This article contains supplementary material online at https://doi.org/10.2337/figshare.27010903.

Acknowledgments. The authors thank Elena Feinstein (Quark Pharmaceuticals) for permission to use the REDD1 KO mice and David Williamson (Penn State Harrisburg) for the use of the REDD1fl/fl mice. The authors are very grateful to Ellen Mullady and Gretchen Snavely, of the Department of Comparative Medicine at Penn State College of Medicine, for their preparation of histology sections.

Funding. This research was supported by grants from the American Diabetes Association (grant no. 11-23-PDF-84), Children’s Miracle Network Trainee Research Award, the Judy S. Finkelstein Memorial Student Research Award, and the Penn State College of Medicine’s Comprehensive Health Studies Program (to S.S.), as well as National Institutes of Health National Eye Institute grant R01 EY032879, and an Innovative Award 1-INO-2024-1538-A-N from JDRF (to M.D.D.).

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. S.S., E.I.Y., A.L.T., H.C., and Y.Z. contributed to the investigation. S.S., A.L.T., H.C., Y.Z., and M.D.D. contributed to methodology. S.S., D.L.G., S.R.K., and M.D.D. conceptualized the study. S.S., D.L.G., S.R.K., and M.D.D. reviewed and edited the manuscript. S.S. and M.D.D. curated the data and contributed to formal analysis, funding acquisition, visualization, and writing the manuscript. S.R.K. and M.D.D. provided resources. S.R.K. and M.D.D. supervised the study. M.D.D. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and accuracy of the data analysis.

1.
Pagtalunan
ME
,
Miller
PL
,
Jumping-Eagle
S
, et al
.
Podocyte loss and progressive glomerular injury in type II diabetes
.
J Clin Invest
1997
;
99
:
342
348
2.
Steffes
MW
,
Schmidt
D
,
McCrery
R
,
Basgen
JM
,
International Diabetic Nephropathy Study Group
.
Glomerular cell number in normal subjects and in type 1 diabetic patients
.
Kidney Int
2001
;
59
:
2104
2113
3.
Sunilkumar
S
,
Yerlikaya
EI
,
Toro
AL
, et al
.
REDD1 ablation attenuates the development of renal complications in diabetic mice
.
Diabetes
2022
;
71
:
2412
2425
4.
Lee
HS
,
Suh
JY
,
Kang
B-C
,
Lee
E
.
Lipotoxicity dysregulates the immunoproteasome in podocytes and kidneys in type 2 diabetes
.
Am J Physiol Renal Physiol
2021
;
320
:
F548
F558
5.
Mundel
P
,
Shankland
SJ
.
Podocyte biology and response to injury
.
J Am Soc Nephrol
2002
;
13
:
3005
3015
6.
Huber
TB
,
Benzing
T
.
The slit diaphragm: a signaling platform to regulate podocyte function
.
Curr Opin Nephrol Hypertens
2005
;
14
:
211
216
7.
Li
JJ
,
Kwak
SJ
,
Jung
DS
, et al
.
Podocyte biology in diabetic nephropathy
.
Kidney Int Suppl
2007
;
72
:
S36
S42
8.
Eid
AA
,
Gorin
Y
,
Fagg
BM
, et al
.
Mechanisms of podocyte injury in diabetes: role of cytochrome P450 and NADPH oxidases
.
Diabetes
2009
;
58
:
1201
1211
9.
Hunt
JL
,
Pollak
MR
,
Denker
BM
.
Cultured podocytes establish a size-selective barrier regulated by specific signaling pathways and demonstrate synchronized barrier assembly in a calcium switch model of junction formation
.
J Am Soc Nephrol
2005
;
16
:
1593
1602
10.
Yang
H
,
Zhao
B
,
Liao
C
, et al
.
High glucose-induced apoptosis in cultured podocytes involves TRPC6-dependent calcium entry via the RhoA/ROCK pathway
.
Biochem Biophys Res Commun
2013
;
434
:
394
400
11.
Möller
CC
,
Wei
C
,
Altintas
MM
, et al
.
Induction of TRPC6 channel in acquired forms of proteinuric kidney disease
.
J Am Soc Nephrol
2007
;
18
:
29
36
12.
Staruschenko
A
,
Spires
D
,
Palygin
O
.
Role of TRPC6 in progression of diabetic kidney disease
.
Curr Hypertens Rep
2019
;
21
:
48
13.
Wang
Y
,
Ding
M
,
Chaudhari
S
, et al
.
Nuclear factor κB mediates suppression of canonical transient receptor potential 6 expression by reactive oxygen species and protein kinase C in kidney cells
.
J Biol Chem
2013
;
288
:
12852
12865
14.
Wang
Y
,
Li
N
,
Wang
Y
, et al
.
NF-κB/p65 competes with peroxisome proliferator-activated receptor gamma for transient receptor potential channel 6 in hypoxia-induced human pulmonary arterial smooth muscle cells
.
Front Cell Dev Biol
2021
;
9
:
656625
15.
Sunilkumar
S
,
Dennis
MD
.
REDD1 is a promising therapeutic target to combat the development of diabetes complications: a report on research supported by Pathway to Stop Diabetes
.
Diabetes
2024
;
73
:
1553
1562
16.
Mu
L
,
Chen
N
,
Chen
Y
, et al
.
Blocking REDD1/TXNIP complex ameliorates HG-induced renal tubular epithelial cell apoptosis and EMT through repressing oxidative stress
.
Int J Endocrinol
2022
;
2022
:
6073911
17.
Wang
X
,
Yang
J
,
Wang
W
,
Li
Y
,
Yang
Y
.
Decreasing REDD1 expression protects against high glucose-induced apoptosis, oxidative stress and inflammatory injury in podocytes through regulation of the AKT/GSK-3β/Nrf2 pathway
.
Immunopharmacol Immunotoxicol
2023
;
45
:
527
538
18.
Sunilkumar
S
,
Dennis
MD
.
FR-PO366: Stress response protein REDD1 sustains the chronic inflammatory response in diabetic nephropathy
.
J Am Soc Nephrol
2023
;
34
:
504
504
19.
Brafman
A
,
Mett
I
,
Shafir
M
, et al
.
Inhibition of oxygen-induced retinopathy in RTP801-deficient mice
.
Invest Ophthalmol Vis Sci
2004
;
45
:
3796
3805
20.
Miller
WP
,
Toro
AL
,
Sunilkumar
S
, et al
.
Müller glial expression of REDD1 is required for retinal neurodegeneration and visual dysfunction in diabetic mice
.
Diabetes
2022
;
71
:
1051
1062
21.
Rangan
GK
,
Tesch
GH
.
Quantification of renal pathology by image analysis
.
Nephrology (Carlton)
2007
;
12
:
553
558
22.
Uil
M
,
Scantlebery
AML
,
Butter
LM
, et al
.
Combining streptozotocin and unilateral nephrectomy is an effective method for inducing experimental diabetic nephropathy in the 'resistant' C57Bl/6J mouse strain
.
Sci Rep
2018
;
8
:
5542
23.
van den Berg
JG
,
van den Bergh Weerman
MA
,
Assmann
KJM
,
Weening
JJ
,
Florquin
S
.
Podocyte foot process effacement is not correlated with the level of proteinuria in human glomerulopathies
.
Kidney Int
2004
;
66
:
1901
1906
24.
Wang
H
,
Sheng
J
,
He
H
, et al
.
A simple and highly purified method for isolation of glomeruli from the mouse kidney
.
Am J Physiol Renal Physiol
2019
;
317
:
F1217
F1223
25.
Hsu
H-H
,
Hoffmann
S
,
Endlich
N
, et al
.
Mechanisms of angiotensin II signaling on cytoskeleton of podocytes
.
J Mol Med (Berl)
2008
;
86
:
1379
1394
26.
Zhou
Y
,
Cai
X
,
Loktionova
NA
, et al
.
The STIM1-binding site nexus remotely controls Orai1 channel gating
.
Nat Commun
2016
;
7
:
13725
27.
Blaine
J
,
Dylewski
J
.
Regulation of the actin cytoskeleton in podocytes
.
Cells
2020
;
9
:
1700
28.
Benzing
T
,
Salant
D
.
Insights into glomerular filtration and albuminuria
.
N Engl J Med
2021
;
384
:
1437
1446
29.
Miao
Z
,
Balzer
MS
,
Ma
Z
, et al
.
Single cell regulatory landscape of the mouse kidney highlights cellular differentiation programs and disease targets
.
Nat Commun
2021
;
12
:
2277
30.
Moeller
MJ
,
Sanden
SK
,
Soofi
A
,
Wiggins
RC
,
Holzman
LB
.
Podocyte-specific expression of cre recombinase in transgenic mice
.
Genesis
2003
;
35
:
39
42
31.
Balkawade
RS
,
Chen
C
,
Crowley
MR
, et al
.
Podocyte-specific expression of Cre recombinase promotes glomerular basement membrane thickening
.
Am J Physiol Renal Physiol
2019
;
316
:
F1026
F1040
32.
Wei
Y
,
Gao
X
,
Li
A
,
Liang
M
,
Jiang
Z
.
Single-nucleus transcriptomic analysis reveals important cell cross-talk in diabetic kidney disease
.
Front Med (Lausanne)
2021
;
8
:
657956
33.
Qi
Z
,
Fujita
H
,
Jin
J
, et al
.
Characterization of susceptibility of inbred mouse strains to diabetic nephropathy
.
Diabetes
2005
;
54
:
2628
2637
34.
Gurley
SB
,
Clare
SE
,
Snow
KP
,
Hu
A
,
Meyer
TW
,
Coffman
TM
.
Impact of genetic background on nephropathy in diabetic mice
.
Am J Physiol Renal Physiol
2006
;
290
:
F214
F222
35.
Siu
B
,
Saha
J
,
Smoyer
WE
,
Sullivan
KA
,
Brosius
FC
.
Reduction in podocyte density as a pathologic feature in early diabetic nephropathy in rodents: Prevention by lipoic acid treatment
.
BMC Nephrol
2006
;
7
:
6
36.
Reiser
J
,
Polu
KR
,
Möller
CC
, et al
.
TRPC6 is a glomerular slit diaphragm-associated channel required for normal renal function
.
Nat Genet
2005
;
37
:
739
744
37.
Ilatovskaya
DV
,
Blass
G
,
Palygin
O
, et al
.
A NOX4/TRPC6 pathway in podocyte calcium regulation and renal damage in diabetic kidney disease
.
J Am Soc Nephrol
2018
;
29
:
1917
1927
38.
Liu
B-C
,
Song
X
,
Lu
X-Y
, et al
.
High glucose induces podocyte apoptosis by stimulating TRPC6 via elevation of reactive oxygen species
.
Biochim Biophys Acta
2013
;
1833
:
1434
1442
39.
Sunilkumar
S
,
Toro
AL
,
McCurry
CM
, et al
.
Stress response protein REDD1 promotes diabetes-induced retinal inflammation by sustaining canonical NF-κB signaling
.
J Biol Chem
2022
;
298
:
102638
40.
Sunilkumar
S
,
VanCleave
AM
,
McCurry
CM
, et al
.
REDD1-dependent GSK3β dephosphorylation promotes NF-κB activation and macrophage infiltration in the retina of diabetic mice
.
J Biol Chem
2023
;
299
:
104991
41.
McCurry
CM
,
Sunilkumar
S
,
Subrahmanian
SM
, et al
.
NLRP3 inflammasome priming in the retina of diabetic mice requires REDD1-dependent activation of GSK3β
.
Invest Ophthalmol Vis Sci
2024
;
65
:
34
42.
Lee
D-K
,
Kim
T
,
Byeon
J
, et al
.
REDD1 promotes obesity-induced metabolic dysfunction via atypical NF-κB activation
.
Nat Commun
2022
;
13
:
6303
43.
Stevens
SA
,
Sunilkumar
S
,
Subrahmanian
SM
, et al
.
REDD1 deletion suppresses NF-κB signaling in cardiomyocytes and prevents deficits in cardiac function in diabetic mice
.
Int J Mol Sci
2024
;
25
:
6461
44.
Graham
S
,
Ding
M
,
Ding
Y
, et al
.
Canonical transient receptor potential 6 (TRPC6), a redox-regulated cation channel
.
J Biol Chem
2010
;
285
:
23466
23476
45.
Kim
EY
,
Anderson
M
,
Wilson
C
,
Hagmann
H
,
Benzing
T
,
Dryer
SE
.
NOX2 interacts with podocyte TRPC6 channels and contributes to their activation by diacylglycerol: essential role of podocin in formation of this complex
.
Am J Physiol Cell Physiol
2013
;
305
:
C960
971
46.
Anderson
M
,
Roshanravan
H
,
Khine
J
,
Dryer
SE
.
Angiotensin II activation of TRPC6 channels in rat podocytes requires generation of reactive oxygen species
.
J Cell Physiol
2014
;
229
:
434
442
47.
Ding
Y
,
Winters
A
,
Ding
M
, et al
.
Reactive oxygen species-mediated TRPC6 protein activation in vascular myocytes, a mechanism for vasoconstrictor-regulated vascular tone
.
J Biol Chem
2011
;
286
:
31799
31809
48.
Miller
WP
,
Sunilkumar
S
,
Dennis
MD
.
The stress response protein REDD1 as a causal factor for oxidative stress in diabetic retinopathy
.
Free Radic Biol Med
2021
;
165
:
127
136
49.
Nguyen
QD
,
Schachar
RA
,
Nduaka
CI
, et al.;
MONET Clinical Study Group
.
Evaluation of the siRNA PF-04523655 versus ranibizumab for the treatment of neovascular age-related macular degeneration (MONET Study)
.
Ophthalmology
2012
;
119
:
1867
1873
50.
Nguyen
QD
,
Schachar
RA
,
Nduaka
CI
, et al.;
DEGAS Clinical Study Group
.
Dose-ranging evaluation of intravitreal siRNA PF-04523655 for diabetic macular edema (the DEGAS study)
.
Invest Ophthalmol Vis Sci
2012
;
53
:
7666
7674
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/journals/pages/license.