Transforming growth factor-β/Smad3 signaling plays an important role in diabetic nephropathy, but its underlying working mechanism remains largely unexplored. The current study uncovered the pathogenic role and underlying mechanism of a novel Smad3-dependent long noncoding RNA (lncRNA) (LRNA9884) in type 2 diabetic nephropathy (T2DN). We found that LRNA9884 was significantly upregulated in the diabetic kidney of db/db mice at the age of 8 weeks preceding the onset of microalbuminuria and was associated with the progression of diabetic renal injury. LRNA9884 was induced by advanced glycation end products and tightly regulated by Smad3, and its levels were significantly blunted in db/db mice and cells lacking Smad3. More importantly, kidney-specific silencing of LRNA9884 effectively attenuated diabetic kidney injury in db/db mice, as shown by the reduction of histological injury, albuminuria excretion, and serum creatinine. Mechanistically, we identified that LRNA9884 promoted renal inflammation-driven T2DN by triggering MCP-1 production at the transcriptional level, and its direct binding significantly enhanced the promoter activity of MCP-1. Thus, LRNA9884 is a novel Smad3-dependent lncRNA that is highly expressed in db/db mice associated with T2DN development. Targeting of LRNA9884 effectively blocked MCP-1–dependent renal inflammation, therefore suppressing the progressive diabetic renal injury in db/db mice. This study reveals that LRNA9884 may be a novel and precision therapeutic target for T2DN in the future.

Type 2 diabetes is a major metabolic disorder that causes a profound medical and socioeconomic burden worldwide (1). Type 2 diabetes is associated with a series of complications on multiple organ systems, where diabetic nephropathy (DN) is the most severe complication of diabetes and is a leading cause of end-stage kidney disease (2). The pathological features of DN include diffuse glomerular basement membrane thickening, mesangial expansion, podocyte reduction, and renal fibrosis (3). Increasing evidence demonstrates that low-grade renal inflammation is a key mechanism leading to diabetes and diabetic complications (4).

Emerging evidence shows that long noncoding RNAs (lncRNAs), defined as RNAs >200 nucleotides that do not encode any protein, play an important role in the development of type 2 diabetic nephropathy (T2DN) (5,6). It has been shown that an lncRNA transcript (lncMGC) is increased in the glomeruli of mouse models of DN in response to endoplasmic reticulum stress and that targeting this lncRNA can protect against early DN (7). In addition, lncRNA CYP4B1-PS1-001 protects against the progression of T2DN via inhibiting proliferation and fibrosis of mesangial cells during the early stage of DN, and lncRNA metastasis associated lung adenocarcinoma transcript 1 (MALAT1) is dysregulated in DN (8,9). A recent study also found that an lncRNA taurine-upregulated gene 1 (Tug1) is reduced in diabetic kidney and that overexpression of Tug1 largely improves mitochondrial bioenergetics in the podocytes of diabetic mice via a peroxisome proliferator–activated receptor-γ coactivator-1a–dependent mechanism (10). Nevertheless, the potential roles and working mechanisms of lncRNAs in DN are still largely unknown (11).

By using high-throughput RNA sequencing, we uncovered the involvement of lncRNAs in renal diseases on both anti- glomerular basement membrane (GBM) and unilateral ureteric obstruction (UUO)-induced mouse kidney disease models and identified 21 novel Smad3-dependent lncRNAs participating in the renal pathogenesis (12). Among these lncRNAs, a novel Smad3-dependent lncRNA, Erbb4-IR, is highly upregulated in the UUO kidney and T2DN with progressive renal fibrosis, and knockdown this lncRNA can protect kidneys from both UUO and diabetic injury (13,14). In the current study, we also found that a potential lncRNA transcript LRNA9884 was the highest expressed Smad3-dependent lncRNAs in the kidneys of db/db mice compared with the db/m mice as well as the Erbb4-IR level. We therefore further elucidated its potential pathogenic role and underlying mechanism in T2DN.

Animal Model

Male db/db and db/m mice were purchased from the Chinese University of Hong Kong Laboratory Animal Services Centre. All mice were fed in a standard animal house with 12-h/12-h light/dark cycle and were sacrificed by intraperitoneal injection of ketamine/xylene. Smad3 knockout (KO) db/db mice were generated by crossing heterozygous db/m with heterozygous Smad3+/− (C57BL/6 background), as previously described (14). All studies and experimental procedures were approved by the Chinese University of Hong Kong Animal Experimentation Ethics Committee, and the experimental methods were performed in accordance with the approved guidelines.

Real-Time PCR Analysis

Total RNA was isolated from the cultured cells and renal cortex of kidney tissues using Trizol (Invitrogen, Carlsbad, CA) according to the manufacturer’s instructions. Real-time PCR was performed by SYBR Green Supermix using the CFX96 PCR System (Bio-Rad, Hercules, CA). The primers used in this study, including mouse collagen I, fibronectin, MCP-1, and β-actin, have been previously described (1518). Other primers include LRNA9884 forward 5′- ACCACAGCAGATACCCAAGC-3′ and reverse 5′- CAGCAAGCTCCTTTTTCCAC-3′. The relative level of the detected gene was normalized with the internal control β-actin. Relative changes in target mRNA expression were determined by using the 2−ΔΔCT method.

Rapid Amplification of cDNA Ends

The full length sequence of LRNA9884 was obtained by conducting rapid amplification of cDNA 5′ and 3′ ends (RACE) with SMARTer RACE 5′/3′ Kit (Clontech, Mountain View, CA), as previously described (13). In brief, the cDNA generated from the total RNA of diabetic injured kidney from the 20-week-old db/db mice was used as the template for RACE PCR according to the user’s manual. The full length of LRNA9884 cDNA was synthesized by PCR with the forward primer 5′- AAAACAGGAGCAGGAAGAATTGGG-3′ and reverse primer 5′- TTGAACTCTGGTCTTCAGAC-3′.

Bioinformatics Analysis

Genomic location of the LRNA9884 was identified by Basic Local Alignment Search Tool (BLAST) (https://blast.ncbi.nlm.nih.gov/Blast.cgi and http://www.genome.ucsc.edu/). Smad3 binding site on the LRNA9884 genomic sequence was identified by the Evolutionary Conserved Regions (ECR) browser (https://ecrbrowser.dcode.org/), as reported in our previous study (12). The protein-coding potential of the LRNA9884 sequence was evaluated by two widely used computational programs: coding potential calculator (CPC) (http://cpc.cbi.pku.edu.cn/programs/run_cpc.jsp) and the coding potential assessment tool (CPAT) (http://lilab.research.bcm.edu/cpat/index.php) (19,20). For evaluation in CPC, transcripts with scores higher than 1 are predicted to be “coding,” lower than −1 are “noncoding,” and between −1 and 1 are classified as “weak noncoding” (−1, 0) or “weak coding” (0, 1). For identifying the direct binding site of LRNA9884 on the MCP-1 promoter, the sequence of LRNA9884 from RACE and the promoter region of MCP-1 from the Eukaryotic Promoter Database (https://epd.vital-it.ch/index.php) were analyzed by IntaRNA tool on the Freiburg RNA Tools website using the default settings (http://rna.informatik.uni-freiburg.de/IntaRNA/Input.jsp) (21), followed by dual-luciferase reporter assay for validating their potential RNA-DNA interaction as in our previous work (13,14).

In Situ Hybridization

The expression level of LRNA9884 in the kidney was detected by using in situ hybridization (ISH), as previously described (14). The kidney sections (4 μm) were fixed in 4% (w/v) paraformaldehyde with 1% (v/v) DMSO. After rehydration, we use a permeabilization step by using proteinase K. Probe for ISH is carried with a locked nucleic acid–digoxigenin labeled LRNA9884 probe (5′-ACTTGAAGGGTCCAGAAGAGAT-3′) (Exiqon, Vedbaek, Denmark) or negative control scramble probe (5′-GTGTAACACGTCTATACGCCCA-3′) (Exiqon) in a humid chamber at 54°C for 2 h. The sections were washed and incubated with anti-digoxigenin antibody conjugated to alkaline phosphatase (Roche Diagnostics, Indianapolis, IN) overnight at 4°C and developed with phosphate/nitroblue tetrazolium (Sigma-Aldrich, St. Louis, MO) (22).

Fluorescence ISH and Immunofluorescence Assay

Fluorescence ISH (FISH) with immunofluorescence assay was performed with a Cy3-conjugated LRAN9884 probe 5′-ACTTGAAGGGTCCAGAAGAGAT-3′ and scramble control 5′-GTGTAACACGTCTATACGCCCA-3′ (GenePharma, Shanghai, P.R. China) by using a FISH kit (C10910; RiboBio, Guangzhou, P.R. China) according to the manufacturer’s instruction. Immunofluorescence assay was followed by FISH, and FITC-conjugated Pan Cytokeratin (53-9003-82; eBioscience, San Diego, CA) and nephrin (ab58968; Abcam, Cambridge, U.K.) were the primary antibodies. All stained samples were imaged under a fluorescence microscope (Axio Observer.Z1; Carl Zeiss, Oberkochen, Germany) (14).

Cell Culture

Immortalized murine kidney proximal tubular epithelial cells (mTECs) (a gift from Dr. Jeffrey B. Kopp, National Institutes of Health) were cultured in DMEM/F-12 (Gibco, Carlsbad, CA), supplemented with 10% FBS (Gibco) and 1% antibiotic/antimycotic solution (Life Technologies, Grand Island, NY) (13,15,16). The murine SV40 transfected mesangial cell line SV40 MES 13 (mMC) (ATCC, Manassas, VA) was maintained in a 3:1 mixture of DMEM and Ham’s F-12 medium containing 5% FBS, and 1% antibiotic/antimycotic solution (Life Technologies) (14). Mouse embryonic fibroblasts (MEFs) lacking Smad3 or Smad2 were generated and characterized as described previously (23), and were cultured in DMEM/F-12 medium (Gibco), as previously described (24). Advanced glycation end products (AGE), at a dose of 50 μg/mL (Cat# A8426; Sigma-Aldrich), was added to induce responses, which was previously dissolved in endotoxin-free 1 × PBS. The composition of AGE includes protein and 95% biuret, and specific of AGE is bovine, their activity on Smad3 activation and MCP-1 expression were validated as position control. To inhibit Smad3 activity, cells were pretreated with the Smad3 inhibitor SIS3 (S0447; Sigma-Aldrich) at a dose-dependent pattern of 1–4 μmol/L for 2 h before AGE stimulation.

Chromatin Immunoprecipitation Analysis

Chromatin immunoprecipitation (ChIP) was performed with the SimpleChIP Enzymatic Chromatin IP Kit (magnetic beads) (#9003; Cell Signaling, Danvers, MA), as previously described (15,18,25). Immunoprecipitation was performed with the antibody against Smad3 (#9513; Cell Signaling) or a normal IgG as a negative control. Precipitated DNA fragments were detected by PCR using a specific primer of promoter region of LRNA9884: forward 5′-TCGCCTCCAGTTGTCTTTCT-3′, reverse 5′-CCTGAAGGACAGCCACTCTC-3′.

Ultrasound-Mediated Gene Transfer-Mediated LRNA9884 Knockdown

Groups of eight male mice were used: db/db mice were treated with pSuper.puro vector (Oligoengine, Seattle WA) empty vector (EV) control or plasmid overexpressing shRNA sequence targeting LRNA9884 (shRNA) (5′-GATCCGACCACAGCAGATACCCAAGCttcaagagaGCTTGGGTATCTGCTGTGGTCTTTTTTGAATTCA-3′) via ultrasound-mediated gene transfer method from the age of week 12 and sacrificed at week 20. To maintain the transgene expression levels, LRNA9884 shRNA expressing plasmids were transfected at age 12, 15, and 18 weeks, as previously described (13,14). In brief, mice received the mixed solution (400 μL/mouse) containing the LRNA9884 shRNA-pSuper.puro vector or pSuper.puro EV (200 μg/mouse) and lipid microbubbles (Sonovue; Bracco, Milan, Italy) at a ratio of 1:1 (v/v) via the tail vein injection, then immediately using an ultrasound transducer (Therasonic; Electro Medical Supplies, Wantage, U.K.) directly placed on the skin of the back against the left kidney with a pulse-wave output of 1 MHz at 2 W/cm2 for a total of 5 min on each side, as previously described (1316).

Histology and Immunohistochemistry

Kidneys sections were fixed in 4% paraformaldehyde, and stained with periodic acid-Schiff (PAS), periodic acid-silver methenamine (PASM), and Masson trichrome staining methods. Immunohistochemistry was performed in 4-μm paraffin sections by using a microwave-based antigen-retrieval technique. The antibodies used in this study included F4/80 (MCA497; Serotec, Oxford, U.K.), interleukin-1β (IL-1β) (sc-7884; Santa Cruz Biotechnology, Dallas, TX), MCP-1 (sc-1785; Santa Cruz Biotechnology), and tumor necrosis factor-α (TNF-α) (sc-1351; Santa Cruz). After immunostaining, sections were counterstained with hematoxylin. The mesangial matrix index was quantified using Image-Pro Plus software (Media Cybernetics, Bethesda, MD) by PAS staining in 10 random fields (original magnification ×200), as previously described (13). Glomerulosclerosis was quantified by Masson trichrome staining, where 20 glomeruli were randomly selected from each section, and positive signals within the selected glomerulus were highlighted, measured, and represented as the percentage positive area of the entire glomerulus. The percentage of positive staining areas of MCP-1, IL-1β, and TNF-α expression was quantified using the same software in 10 consecutive fields, whereas positive cells for F4/80+ cells were counted under the power field of microscope in 10 random areas of kidney tissues using a 0.25-mm2 graticule fitted in the eyepiece of the microscope and expressed as cells/mm2 (13,14,26).

Renal Function Measurement

Mice were placed in metabolic cages for collection of 24-h urinary samples every 4 weeks from the age of 8 weeks. Urinary microalbumin was measured by competitive ELISA according to the manufacturer’s instructions (Exocell, Philadelphia, PA). Urinary and serum creatinine was measured by an enzymatic kit (Stanbio Laboratories, Boerne, TX). Urinary albumin excretion was expressed as total urinary albumin–to–creatinine ratio (μg/mg), as previously reported (26).

Fasting Blood Glucose

Blood glucose levels were measured every 4 weeks by Accu-Chek glucose meter (Roche Diagnostics, Basel, Switzerland) after the mice were fasted for 6 h, as recommended by the Animal Models of Diabetic Complications Consortium.

Transfection of siRNA and Plasmid

The mTECs were transfected with 100 nmol/L LRNA9884 siRNA (sense 5′-GACCACAGCAGAUACCCAAGC-3′, antisense 5′-GCUUGGGUAUCUGCUGUGGUC-5′) or nonsense control (NC; sense 5′-UUCUCCGAACGUGUCACGUTT-3′, antisense 3′-ACGUGACACGUUCGGAGAATT-5′) using Lipofectamine RNAiMAX reagent (Invitrogen) according to the manufacturer’s instructions as previously reported (27), then stimulated with AGE. In addition, the mTECs were also transfected with 0.5 μg/mL pcDNA3.1 EV or plasmid containing LRNA9884 full-length RNA sequence, as shown in Supplementary Fig. 1 (LRNA9884; constructed by PharmaGene, Shanghai, China) using Lipofectamine 2000 (Invitrogen) according to the manufacturer’s manual, and the culture.

Western Blot Analysis

Protein from renal cortex and cultured cells was extracted using radio immunoprecipitation assay (RIPA) lysis buffer. Western blot analysis was performed as previously described (28,29). In brief, after nonspecific binding was blocked with 5% BSA, nitrocellulose membranes were incubated overnight at 4°C with primary antibodies against phosphorylated-Smad3 (Cell Signaling Technology, Danvers, MA) and Smad3 (Cell Signaling Technology), collagens I (1310-01; Southern Tech, Birmingham, AL), fibronectin (sc-8422; Santa Cruz Biotechnology), and β-actin (sc-47778; Santa Cruz Biotechnology), followed by IRDye800-conjugated secondary antibodies (Rockland Immunochemicals, Gilbertsville, PA). Signals were detected using the LI-COR/Odyssey infrared image system (LI-COR Biosciences, Lincoln, NE), followed by quantitative analysis using the Image J program (https://imagej.nih.gov/ij/?) (13,18). The ratio for the protein examined was normalized against β-actin and expressed as the mean ± SEM.

ELISA

Medium from stimulated mTECs was collected to detect the cytokine production using an ELISA kit. MCP-1 was measured with Quantikine ELISA Kit (R&D Systems, Minneapolis, MN) according to the product protocols.

Dual-Luciferase Reporter Assay

The pcDNA3.1+ plasmids containing LRNA9884 full-length sequence with (LRNA9884) or without (LRNA9884-mutant) predicted the binding site on the MCP-1 promoter; and pGL3-basic reporter plasmids containing MCP-1 promoter sequence with (pGL3–MCP-1) or without predicted LRNA9884 binding site (pGL3–MCP-1–mutant) were constructed by Biowit Technologies Ltd., China (13,14). The luciferase reporter assay was performed by Landbiology (Guangzhou, China) by using dual luciferase reporter assay kit (Promega, Madison, WI) as our previous studies (24). The empty vectors (pGL3-basic, pcDNA3.1+), or MCP-1 reporter plasmids (pGL3–MCP-1, pGL3–MCP-1–mutant), or LRNA9884-overexpressing pcDNA3.1+ plasmids (LRNA9884, LRNA9884-mutant) were cotransfected into mTECs. Luciferase activities were measured at 48 h according to the manufacturer’s instructions by GloMax-Multi Detection System (Promega). The reporter activity was represented by the ratio of the firefly luciferase activity (M1) to the Renilla luciferase activity (M2) as M1/M2, and shown as mean ± SEM fold induction of luciferase in three independent experiments.

Statistical Analysis

All of the data are expressed as mean ± SEM. Statistical analyses were performed with one-way ANOVA, followed by Newman-Keuls multiple comparison from GraphPad Prism 5.0 (GraphPad Software, La Jolla, CA). In addition, a repeated-analysis ANOVA was used for albumin excretion, body weight, and fasting blood glucose analysis.

LRNA9884 Is a Novel Smad3-Dependent lncRNA and Is Highly Expressed and Associated With Progression of Renal Injury in db/db Mice

Among all previously identified Smad3-depedent lncRNAs (11), real-time PCR analysis revealed that a potential lncRNA transcript (na_9884) was highly expressed in the diabetic kidney of db/db mice (Fig. 1A). More importantly, expression of na_9884 progressively increased in the diabetic kidneys of db/db mice compared with db/m since the age of 8 weeks (Fig. 1B). We then further characterized this novel lncRNA by obtaining its full-length sequence via RACE (Supplementary Fig. 1). Our result identified that na_9884 is 508 nucleotides in full length, which locates on murine chromosome 5 (Chr5: 52973035–52992344) and covers the whole genomic sequence of an lncRNA transcript 5033403H07Rik (Fig. 1C and D). By using the CPC and CPAT analysis, we confirmed that na_9884 scores −1.01155 and 0, respectively, as an RNA without protein-coding capacity (Fig. 1E). Thus, we named this novel lncRNA as LRNA9884. By ISH, we further observed that LRNA9884 is highly expressed in the nucleus of renal residential cells in the diabetic kidney of 8-week-old db/db mice and further upregulated in 20-week-old db/db mice (Fig. 1F).

Figure 1

Characterization of LRNA9884 in kidney. A: Real-time PCR shows expression of 21 common lncRNAs in db/db and db/m mice (n = 8 in each group). B: Real-time PCR detects the upregulation of na_9884 in the kidney during progression of db/db mice from the age of 8 weeks. C: A full length of na_9884 (508 base pairs [bp], named as LRNA9884) amplified by PCR is detected by electrophoresis in 1% agarose gel. D: Gene location of LRNA9884 in chromosome 5 of the mouse genome. E: Scores calculated by using the CPC and CPAT analysis. UTR, untranslated region. F: ISH detects LRNA9884 expression (nuclear pattern, presumably by glomerular [g] and tubular cells) at the age of 8 weeks and 20 weeks in the kidneys of db/db and db/m mice (original magnification ×400). Each bar represents the mean ± SEM for groups of eight mice. *P < 0.05, **P < 0.01, ***P < 0.001 vs. db/m mice.

Figure 1

Characterization of LRNA9884 in kidney. A: Real-time PCR shows expression of 21 common lncRNAs in db/db and db/m mice (n = 8 in each group). B: Real-time PCR detects the upregulation of na_9884 in the kidney during progression of db/db mice from the age of 8 weeks. C: A full length of na_9884 (508 base pairs [bp], named as LRNA9884) amplified by PCR is detected by electrophoresis in 1% agarose gel. D: Gene location of LRNA9884 in chromosome 5 of the mouse genome. E: Scores calculated by using the CPC and CPAT analysis. UTR, untranslated region. F: ISH detects LRNA9884 expression (nuclear pattern, presumably by glomerular [g] and tubular cells) at the age of 8 weeks and 20 weeks in the kidneys of db/db and db/m mice (original magnification ×400). Each bar represents the mean ± SEM for groups of eight mice. *P < 0.05, **P < 0.01, ***P < 0.001 vs. db/m mice.

Close modal

LRNA9884 Is Markedly Induced in Renal Tubular Epithelial and Mesangial Cells Under Diabetic Condition

The LRNA9884 expression was further confirmed by FISH assay with immunofluorescence costaining. Owing to lack of the specific marker in mouse mesangial cells, the epithelial cell marker keratin and podocyte marker nephrin were detected in kidneys of 20-week-old db/db mice. Results demonstrated that LRNA9884 was largely expressed in the nucleus of epithelial cells and was lacking in podocytes compared with the db/m mice (Fig. 2A), suggesting that LRNA9884 was mainly expressed in epithelial cells and mesangial cells. Aberrant glucose metabolism results in hyperglycemia, and accumulation of various byproducts (e.g., AGE) is suggested to promote renal injury in diabetic kidney disease. Thus, we cultured mTECs and mMCs under high glucose or AGE conditions in vitro. We found that LRNA9884 expression was significantly upregulated in both mTECs and mMCs at 1 h by AGE (50 μg/mL) stimulation in a time- and dose-dependent manner (Fig. 2B–E), whereas its induction was significant at 3 h under high glucose (35 mmol/L) stimulation (Supplementary Fig. 2).

Figure 2

LRNA9884 expression is tightly regulated by AGE in mTECs and mMCs. A: FISH assay with immunofluorescence containing epithelial cell marker keratins (second panels from left) and podocyte marker nephrons (right-most panels) confirmed the cell-specific expression of LRNA9884 (red) with the nucleus (DAPI, blue) in the diabetic injured kidney of 20-week-old db/db mice. Real-time PCR shows that AGE (50 μg/mL) induces LRNA9884 expression in mTECs (B and C) and mMCs (D and E) in a time- and dose-dependent manner. Data represent the mean ± SEM for at least three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001 vs. time-matched control or control group. #P < 0.05 vs. AGE (50 μg/mL) concentration.

Figure 2

LRNA9884 expression is tightly regulated by AGE in mTECs and mMCs. A: FISH assay with immunofluorescence containing epithelial cell marker keratins (second panels from left) and podocyte marker nephrons (right-most panels) confirmed the cell-specific expression of LRNA9884 (red) with the nucleus (DAPI, blue) in the diabetic injured kidney of 20-week-old db/db mice. Real-time PCR shows that AGE (50 μg/mL) induces LRNA9884 expression in mTECs (B and C) and mMCs (D and E) in a time- and dose-dependent manner. Data represent the mean ± SEM for at least three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001 vs. time-matched control or control group. #P < 0.05 vs. AGE (50 μg/mL) concentration.

Close modal

LRNA9884 Expression Is Tightly Regulated by Smad3 Signaling In Vitro and In Vivo

In our previous study, induction of LRNA9884 was associated with chronic kidney disease mouse models in a Smad3-dependent manner as revealed by high-throughput RNA sequencing (12), but its regulatory mechanism in diabetic kidney injury is still unknown. Interestingly, we observed the renal expression of LRNA9884 in 12-week-old db/db mice was largely suppressed after Smad3 KO, as shown by ISH and real-time PCR (Fig. 3A and B). Thus, we further speculated that LRNA9884 may be regulated by Smad3 under diabetic conditions. This was examined in MEFs, in which AGE-induced upregulation of LRNA9884 was also largely suppressed in MEFs, knocking out Smad3 but not Smad2 (Fig. 3C and D and Supplementary Fig. 3A and B). By using the ECR browser, we predicted a conversed Smad3 binding site on the promoter region of LRNA9884 genomic sequence (Fig. 4A). The physical binding of Smad3 protein on the promoter region of the LRNA9884 genomic sequence was dramatically enriched in mTECs by AGE stimulation as demonstrated by ChIP assay (Fig. 4B). Thus deletion (Smad3 KO) or inhibition (SIS3) of Smad3 effectively blocked the AGE-induced LRNA9884 expression in MEFs and mTECs in vitro, (Fig. 4C and D and Supplementary Fig. 3). Thus, our findings clearly demonstrated that expression of LRNA9884 is tightly regulated by Smad3 signaling and serves as a direct Smad3 target gene under diabetic condition in vitro and in vivo.

Figure 3

LRNA9884 expression is positively regulated via Smad3-dependent mechanism in vitro and in vivo. A and B: ISH and real-time PCR detect LRNA9884 is highly expressed in the diabetic kidney (week 12) of S3WT db/db mice. In contrast, expression of renal LRNA9884 in S3KO db/db mice is comparable to normal db/m mice (×400). g, glomerulus. C and D: Real-time PCR analysis shows that AGE induces LRNA9884 expression in Smad3 WT (S3WT) MEF but not in Smad3 KO (S3KO) MEF cells. Each bar represents the mean ± SEM for groups of six mice or three independent experiments. *P < 0.05, **P < 0.01 vs. db/m or time match control group. ###P < 0.001 vs. db/db-S3WT group.

Figure 3

LRNA9884 expression is positively regulated via Smad3-dependent mechanism in vitro and in vivo. A and B: ISH and real-time PCR detect LRNA9884 is highly expressed in the diabetic kidney (week 12) of S3WT db/db mice. In contrast, expression of renal LRNA9884 in S3KO db/db mice is comparable to normal db/m mice (×400). g, glomerulus. C and D: Real-time PCR analysis shows that AGE induces LRNA9884 expression in Smad3 WT (S3WT) MEF but not in Smad3 KO (S3KO) MEF cells. Each bar represents the mean ± SEM for groups of six mice or three independent experiments. *P < 0.05, **P < 0.01 vs. db/m or time match control group. ###P < 0.001 vs. db/db-S3WT group.

Close modal
Figure 4

LRNA9884 expression is transcriptionally induced by AGE via a Smad3-dependent mechanism. A: The evolutionarily conserved region of LRNA9884 in human (hg19) and mouse (mm10) genomes and the predicted binding site by Smad3 analyzed by the ECR Browser (underlined). B: ChIP assay shows that Smad3 physically binds the LRNA9884 promoter in response to AGE (50 μg/mL). C: Real-time PCR analysis shows AGE induces LRNA9884 expression in Smad3 WT MEF but is absent in Smad3KO MEF cells. D: Real-time PCR shows that the addition of SIS3 inhibits AGE (50 μg/mL)-induced LRNA9884 expression in mTECs at 1 h. Each bar represents the mean ± SEM for three independent experiments. **P < 0.01 vs. control group. #P < 0.05, ##P < 0.01, ###P < 0.001 vs. SIS3 (0 μmol/L) concentration in response to AGE (50 μg/mL) or db/db S3WT group.

Figure 4

LRNA9884 expression is transcriptionally induced by AGE via a Smad3-dependent mechanism. A: The evolutionarily conserved region of LRNA9884 in human (hg19) and mouse (mm10) genomes and the predicted binding site by Smad3 analyzed by the ECR Browser (underlined). B: ChIP assay shows that Smad3 physically binds the LRNA9884 promoter in response to AGE (50 μg/mL). C: Real-time PCR analysis shows AGE induces LRNA9884 expression in Smad3 WT MEF but is absent in Smad3KO MEF cells. D: Real-time PCR shows that the addition of SIS3 inhibits AGE (50 μg/mL)-induced LRNA9884 expression in mTECs at 1 h. Each bar represents the mean ± SEM for three independent experiments. **P < 0.01 vs. control group. #P < 0.05, ##P < 0.01, ###P < 0.001 vs. SIS3 (0 μmol/L) concentration in response to AGE (50 μg/mL) or db/db S3WT group.

Close modal

Kidney-Specific Silencing of LRNA9884 Protects Against Diabetic Renal Injury in db/db Mice

Because LRNA9884 induction was highly associated with T2DN progression (Fig. 1B and F), we further investigated its potential role in the pathogenesis of T2DN in db/db mice. EVs or plasmids expressing shRNA specifically against LRNA9884 were transfected into both kidneys of the db/db mice (100 μg/mouse per treatment) by using an ultrasound-microbubble delivery system (9). Treatment with shRNA effectively suppressed expression of renal LRNA9884 in db/db mice compared with the EV-treated 20-week-old control mice according to results of real-time PCR and ISH (Fig. 5A and B). More importantly, inhibition of renal LRNA9884 significantly suppressed renal histological injury, including glomerular matrix deposition and glomerulosclerosis (Fig. 5A, C, and D), and decreased microalbuminuria excretion and serum creatinine (Fig. 5E and F) as determined by PAS, PASM, and Masson trichrome stain and ELISA assays. However, blockade of renal LRNA9884 showed no significant therapeutic effects on the type 2 diabetes phenotype, including body weight and fasting blood glucose (Supplementary Fig. 4), suggesting LRNA9884 promotes type 2 diabetes complications via a kidney-specific mechanism.

Figure 5

Kidney-specific silencing of LRNA9884 improves renal histology and function in db/db mice. A: ISH, PAS staining, PASM staining, and Masson trichrome staining show changes in renal histology after knockdown of LRNA9884 in db/db kidney. B: Real-time PCR shows that ultrasound-microbubble-mediated LRNA9884 shRNA (shRNA) transfer effectively inhibits its expression in db/db kidney at the age of 20 weeks compared with the EV-treated control. C and D: Quantification analysis of mesangial matrix index by PAS staining and glomerulosclerosis by Masson trichrome staining at the age of 20 weeks in kidneys. E and F: Urine microalbuminuria-to-creatinine ratio (UACR) and serum creatinine in db/db mice after knockdown of LRNA9884. Data represent the mean ± SEM for eight mice per group. **P < 0.01, ***P < 0.001 vs. db/m mice; #P < 0.05, ##P < 0.01 vs. db/db mice with EV treatment.

Figure 5

Kidney-specific silencing of LRNA9884 improves renal histology and function in db/db mice. A: ISH, PAS staining, PASM staining, and Masson trichrome staining show changes in renal histology after knockdown of LRNA9884 in db/db kidney. B: Real-time PCR shows that ultrasound-microbubble-mediated LRNA9884 shRNA (shRNA) transfer effectively inhibits its expression in db/db kidney at the age of 20 weeks compared with the EV-treated control. C and D: Quantification analysis of mesangial matrix index by PAS staining and glomerulosclerosis by Masson trichrome staining at the age of 20 weeks in kidneys. E and F: Urine microalbuminuria-to-creatinine ratio (UACR) and serum creatinine in db/db mice after knockdown of LRNA9884. Data represent the mean ± SEM for eight mice per group. **P < 0.01, ***P < 0.001 vs. db/m mice; #P < 0.05, ##P < 0.01 vs. db/db mice with EV treatment.

Close modal

Silencing of LRNA9884 Attenuates Renal Inflammation In Vivo and In Vitro

Immunohistochemistry and real-time PCR detected that a significant reduction of inflammatory markers (i.e., MCP-1, TNF-α, and IL-1β) occurred in the kidney of 20-week-old db/db mice by shRNA-9884 treatment compared with the EV-treated and db/db controls (Fig. 6). To elucidate the detailed mechanism mediated by LRNA9884, we screened Smad3-related downstream inflammatory biomarkers after knockdown LRNA9884 by using real-time PCR. Results showed that AGE-induced inflammation and proinflammation cytokines were downregulated after silencing of LRNA9884 in vitro, among which inhibition of MCP-1 expression by shRNA treatment was the most remarkable one, suggesting that LRNA9884 may trigger MCP-1–dependent renal injury under diabetic condition (Supplementary Fig. 5).

Figure 6

Knockdown of LRNA9884 inhibits inflammatory response in the db/db kidney. A: Immunohistochemistry (IHC) and quantitative analysis of MCP-1, IL-1β, and TNF-α expression (×400). B: Real-time PCR analysis of MCP-1, IL-1β, and TNF-α mRNA expression in mice kidneys. Data represent the mean ± SEM for six mice per group. *P < 0.05, **P < 0.01 vs. db/m mice; #P < 0.05 vs. db/db mice with EV treatment.

Figure 6

Knockdown of LRNA9884 inhibits inflammatory response in the db/db kidney. A: Immunohistochemistry (IHC) and quantitative analysis of MCP-1, IL-1β, and TNF-α expression (×400). B: Real-time PCR analysis of MCP-1, IL-1β, and TNF-α mRNA expression in mice kidneys. Data represent the mean ± SEM for six mice per group. *P < 0.05, **P < 0.01 vs. db/m mice; #P < 0.05 vs. db/db mice with EV treatment.

Close modal

In addition, no significant changes in the AGE-induced renal fibrotic marker expression (i.e., collagen I, fibronectin, and phosphorylated Smad3) in mTECs after effective siRNA-mediated knockdown of LRNA9884 (siLRNA9884) compared with the nonsense-treated control as shown by real-time PCR and Western blot analysis (Fig. 7 and Supplementary Figs. 5 and 6). MCP-1 has a crucial role in renal inflammation via promoting the infiltration of inflammatory leukocytes into injured kidney; therefore, the underlying mechanism of how LRNA9884 regulates MCP-1 production at the transcriptional level was further elucidated in this study, as shown below.

Figure 7

Knockdown of LRNA9884 has no effect on AGE-induced fibrotic responses in mTECs. A: Real-time PCR shows LRNA9884 expression compared with nonsense-treated control (NC) after AGE-induced in mTECs. Real-time PCR shows fibronectin (Fn) (B) and collagen I (Col-I) (C) expression after knockdown of LRNA9884 in mTECs. D: Western blot (WB) analysis shows expression of Fn and Col-I in mTECs. Quantitative analysis of Fn (E) and Col-I (F) in mTECs. Data represent the mean ± SEM for at least three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001 vs. time 0 h. ##P < 0.01 vs. time-matched control group.

Figure 7

Knockdown of LRNA9884 has no effect on AGE-induced fibrotic responses in mTECs. A: Real-time PCR shows LRNA9884 expression compared with nonsense-treated control (NC) after AGE-induced in mTECs. Real-time PCR shows fibronectin (Fn) (B) and collagen I (Col-I) (C) expression after knockdown of LRNA9884 in mTECs. D: Western blot (WB) analysis shows expression of Fn and Col-I in mTECs. Quantitative analysis of Fn (E) and Col-I (F) in mTECs. Data represent the mean ± SEM for at least three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001 vs. time 0 h. ##P < 0.01 vs. time-matched control group.

Close modal

LRNA9884 Directly Triggers MCP-1 Production at the Transcriptional Level, Thereby Promoting Renal Inflammation Under Diabetic Conditions

Mechanistically, we found that LRNA9884 was continuously increased in the nucleus of mesangial and tubular epithelial cells during T2DN progression, which was associated with a marked upregulation of MCP-1 in the diabetic injured kidney (Figs. 1B and 6B). We also found that a potential binding site of LRNA9884 is predicted on the promoter region of the MCP-1 genomic sequence by using the Freiburg RNA Tool (Fig. 8A and Supplementary Fig. 7). To investigate the functional role of LRNA9884, we performed siRNA-mediated knockdown of LRNA9884 on mTECs and found that silencing of LRNA9884 largely inhibited AGE-induced MCP-1 production in mTECs (Fig. 8B and C and Supplementary Fig. 8). Furthermore, overexpression of LRNA9884 could directly upregulate the expression of MCP-1 without AGE stimulation (Fig. 8D and Supplementary Fig. 8). More importantly, direct physical interaction between LRNA9884 and the promoter region of the MCP-1 genomic sequence was clearly demonstrated in vitro by dual-luciferase reporter assay, where overexpression of full-length LRNA9884 significantly enhanced the transcription activity of MCP-1 via direct binding on its promoter region (pGL3–MCP-1–promoter), which was effectively prevented by deletion of the predicted LRNA9884 binding site on the MCP-1 promoter (pGL3–MCP-1–mutant) or the LRNA9884 full-length sequence (pcDNA3.1-LRNA9884-mutant) (Fig. 8E). Therefore, targeting of renal LRNA9884 effectively protected against AGE-driven diabetic kidney injury by blocking renal inflammation via a MCP-1–dependent mechanism. This was further demonstrated by a significant reduction of the kidney-infiltrating macrophages in shRNA-9884–treated diabetic kidney compared with EV-treated and untreated db/db controls (Supplementary Fig. 9). Therefore, LRNA9884 may represent as an effective novel therapeutic target for AGE-driven T2DN.

Figure 8

LRNA9884 directly interacts with MCP-1 in mTECs. A: Predicted binding site of LRNA9884 on the genomic sequence of MCP-1. B: ELISA detects MCP-1 protein expression in cell culture medium. C: Western blot shows MCP-1 protein expression after siRNA-mediated knockdown of LRNA9884 (siLRNA9884) in mTECs. D: Western blot shows that transfection of LRNA9884-expressing pcDNA3.1+ plasmid increased MCP-1 protein expression in mTECs compared with the EV (pcDNA3.1+) control. E: Dual luciferase assay shows the enhanced effect of LRNA9884 on MCP-1 expression via its binding on the MCP-1 promoter region, which is prevented by deletion of the LRNA9884 binding site in MCP-1 promoter (pGL3–mutant–MCP-1) or LRNA9884 sequences (LRNA9884-mutant), respectively. Data represent the mean ± SEM for at least three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001 vs. time 0 h or pGL3-basic group. #P < 0.05, ##P < 0.01 vs. time-matched control or pGL3–MCP-1 group, ^^P < 0.01 vs. LRNA9884 group.

Figure 8

LRNA9884 directly interacts with MCP-1 in mTECs. A: Predicted binding site of LRNA9884 on the genomic sequence of MCP-1. B: ELISA detects MCP-1 protein expression in cell culture medium. C: Western blot shows MCP-1 protein expression after siRNA-mediated knockdown of LRNA9884 (siLRNA9884) in mTECs. D: Western blot shows that transfection of LRNA9884-expressing pcDNA3.1+ plasmid increased MCP-1 protein expression in mTECs compared with the EV (pcDNA3.1+) control. E: Dual luciferase assay shows the enhanced effect of LRNA9884 on MCP-1 expression via its binding on the MCP-1 promoter region, which is prevented by deletion of the LRNA9884 binding site in MCP-1 promoter (pGL3–mutant–MCP-1) or LRNA9884 sequences (LRNA9884-mutant), respectively. Data represent the mean ± SEM for at least three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001 vs. time 0 h or pGL3-basic group. #P < 0.05, ##P < 0.01 vs. time-matched control or pGL3–MCP-1 group, ^^P < 0.01 vs. LRNA9884 group.

Close modal

It is well accepted that DN as a severe diabetic complication is mediated by TGF-β1 via both Smad3-dependent and non–Smad3-dependent signaling pathways (3,30,31). In this study, we uncovered the pathogenic role of a novel Smad3-dependent LRNA9884 in the disease progression of T2DN and identified its molecular mechanism under diabetic conditions in vivo and in vitro. We found that LRNA9884 was a Smad3-dependent lncRNA highly expressed in the diabetic kidneys and by AGE-stimulated mMCs/mTECs of Smad3 wild-type (WT) but not Smad3 KO. We also found that the expression levels of LRNA9884 were correlated with progressive diabetic kidney disease in db/db mice. Further characterization by obtaining its full-length sequence via RACE showed that LRNA9884 is 508 nucleotides in length and locates within 5033403H07Rik at murine chromosome 5 (Chr5: 52973035–52972344). Since LRNA9884 covered the whole genomic sequence of its host gene, we speculated that LRNA9884 is the completed sequence of 5033403H07Rik. Through the use of CPC and CPAT analysis, LRNA9884 was confirmed and classified as a novel noncoding RNA.

We found that expression of LRNA9884 was significantly induced by the metabolic byproduct and hyperglycemia (AGE and high glucose) under diabetic conditions but not by the profibrotic factor TGF-β1 in the cultured renal cells in vitro. Furthermore, it was markedly upregulated in the db/db mice kidneys but not in the UUO kidneys, as previously reported (13), which implied that LRNA9884 is specifically induced under diabetic conditions. However, LRNA9884 has no regulatory role in glucose metabolism, as evidenced by blocking LRNA9884 without altering the fasting blood glucose levels and body weight in db/db mice. The high disease- and tissue-specificity of LRNA9884 encouraged us to further elucidate its therapeutic potential for T2DN.

First, we identified the biological function of LRNA9884 in the development of T2DN and found that knockdown of LRNA9884 effectively inhibited the AGE-induced inflammation markers, including MCP-1, IL-1β, and TNF-α, on cultured mTECs without influencing on the expression of fibrosis markers such as collagen I and fibronectin in vitro. The inflammatory role for LRNA9884 was further confirmed in vivo, where specifically silencing of LRNA9884 in the kidney of db/db mice was capable of inhibiting renal inflammation. Indeed, renal LRNA9884 was significantly upregulated at the age of 8 weeks before the onset of detectable diabetic renal injury, such as microalbuminuria, suggesting that LRNA9884 may be pathogenic and responsible for the early development of T2DN in db/db mice. It is likely that the early upregulation of LRNA9884 may trigger the early development of low-grade renal inflammation rather than renal fibrosis, because inhibition of LRNA9884 expression suppressed renal inflammation in db/db mice and in cultured mTECs without effect on the expression of profibrotic cytokines that mainly exert a role in renal fibrosis during the late progression of T2DN (3,3234). Thus, LRNA9884 may function to trigger the development of low-grade renal inflammation in the early development of T2DN.

Second, we revealed that LRNA9884 was tightly regulated by Smad3 under the diabetic condition. We previously reported that AGE is able to activate the Smad-signaling pathway via both TGF-β–dependent and –independent manners (30,31). In this study, we found that the promoter region of LRNA9884 contained a Smad3-binding site and that deletion of Smad3 dramatically blocked the upregulation of LRNA9884 in the diabetic kidney of db/db mice in vivo and in AGE-stimulated MEFs in vitro, suggesting a positive regulatory role of Smad3 in the induction of LRNA9884 during renal inflammation under diabetic conditions. Interestingly, although LRNA9884 was upregulated in the kidney of Smad3-WT db/db mice, it did not participate in the AGE/Smad3-mediated renal fibrosis because knockdown of LRNA9884 did not influence expression of Col-I and fibronectin in vitro. Thus, LRNA9884 is a downstream mediator of Smad3 specific for promoting renal inflammation under diabetic conditions.

Importantly, we detected that the LRNA9884 may mediate renal inflammation in db/db mice via a MCP-1–dependent mechanism. This was supported by the findings that a physical interaction exists between LRNA9884 and the MCP-1 promoter as identified by ChIP and dual-luciferase reporter assays. Moreover, we also demonstrated that silencing of LRNA9884 dramatically suppressed MCP-1 production in the diabetic kidney of db/db mice and in AGE-stimulated mTECs. The diabetic milieu is a low-grade inflammatory disease, where the kidney-infiltrating macrophages largely promote renal inflammation-driven DN (35). Silencing of LRNA9884 reduced the renal MCP-1 level, which largely suppressed the macrophage infiltration and therefore significantly reduced the renal inflammation-driven kidney injured in db/db mice. All of these findings revealed that MCP-1 should be an important downstream target gene of LRNA9884 responsible for the T2DN development. In this study, we identified the main function of LRNA9884 is promoting tissue inflammation. Nevertheless, the biological function and working mechanism of this novel Smad3-dependent lncRNA is still largely unknown, and an unbiased approach to determine whether LRNA9884 alters the gene expression of more than MCP-1 should be further investigated.

Finally, results from this study also provided evidence that targeting LRNA9884 specifically on the diabetic kidney may represent a novel therapy for T2DN. Consistent with previous reports (7,14), by using a noninvasive ultrasound-microbubble-mediated technique, we were able to effectively silence renal LRNA9884-mediated MCP-1 production in db/db mice, which largely blunted the kidney infiltration of inflammatory leukocytes (e.g., macrophages), thereby suppressing renal inflammation and resulting in improved renal function in the db/db mice with diabetic kidney diseases. It should be noted that although we tested the specificity of siLRNA9884 by BLAST analysis, the siLRNA9884 vector used in our in vivo experiments may have had effects on other genes or transcripts than those targeted.

In conclusion, the current study identifies that LRNA9884 is a Smad3-dependent lncRNA and plays a proinflammatory role in T2DN via an MCP-1–dependent mechanism. Our findings suggest that targeting renal LRNA9884 may represent a novel and precision therapy for AGE-driven diabetic kidney diseases.

Funding. This study was supported by Research Grants Council of Hong Kong (GRF 14106518, 14117418, 14117815, 14121816, 14163317, C7018-16G, TRS T12-402/13N), Health and Medical Research Fund (03140486, 14152321), Direct Grant for Research CUHK (2017.002), Lui Che Woo Institute of Innovative Medicine (CARE), and the Major State Basic Research Development Program of China (2012CB517705).

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

Author Contributions. Y.-y.Z., P.M.-K.T., P.C.-T.T., J.X., X.-R.H., C.Y., R.C.W.M., and H.-Y.L. approved the final version of the manuscript. Y.-y.Z. drafted the original manuscript. Y.-y.Z. and P.M.-K.T. analyzed the data and created the figures. Y.-y.Z., P.M.-K.T., P.C.-T.T., J.X., and X.-R.H. performed experiments. P.M.-K.T. and H.-Y.L. reviewed and edited the writing. X.-R.H., C.Y., and R.C.W.M. supported resources. R.C.W.M. and H.-Y.L. covered the funding. H.-Y.L. designed the study. H.-Y.L. 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 the accuracy of the data analysis.

1.
Zimmet
PZ
,
Magliano
DJ
,
Herman
WH
,
Shaw
JE
.
Diabetes: a 21st century challenge
.
Lancet Diabetes Endocrinol
2014
;
2
:
56
64
[PubMed]
2.
Sharma
K
,
Karl
B
,
Mathew
AV
, et al
.
Metabolomics reveals signature of mitochondrial dysfunction in diabetic kidney disease
.
J Am Soc Nephrol
2013
;
24
:
1901
1912
[PubMed]
3.
Lan
HY
.
Transforming growth factor-β/Smad signalling in diabetic nephropathy
.
Clin Exp Pharmacol Physiol
2012
;
39
:
731
738
[PubMed]
4.
Navarro-González
JF
,
Mora-Fernández
C
,
Muros de Fuentes
M
,
García-Pérez
J
.
Inflammatory molecules and pathways in the pathogenesis of diabetic nephropathy
.
Nat Rev Nephrol
2011
;
7
:
327
340
[PubMed]
5.
Panchapakesan
U
,
Pollock
C
.
Long non-coding RNAs-towards precision medicine in diabetic kidney disease
?
Clin Sci (Lond)
2016
;
130
:
1599
1602
[PubMed]
6.
Long
J
,
Danesh
FR
.
Values and limitations of targeting lncRNAs in diabetic nephropathy
.
Diabetes
2018
;
67
:
552
553
[PubMed]
7.
Kato
M
,
Wang
M
,
Chen
Z
, et al
.
An endoplasmic reticulum stress-regulated lncRNA hosting a microRNA megacluster induces early features of diabetic nephropathy
.
Nat Commun
2016
;
7
:
12864
[PubMed]
8.
Wang
M
,
Wang
S
,
Yao
D
,
Yan
Q
,
Lu
W
.
A novel long non-coding RNA CYP4B1-PS1-001 regulates proliferation and fibrosis in diabetic nephropathy
.
Mol Cell Endocrinol
2016
;
426
:
136
145
[PubMed]
9.
Hu
M
,
Wang
R
,
Li
X
, et al
.
LncRNA MALAT1 is dysregulated in diabetic nephropathy and involved in high glucose-induced podocyte injury via its interplay with β-catenin
.
J Cell Mol Med
2017
;
21
:
2732
2747
[PubMed]
10.
Long
J
,
Badal
SS
,
Ye
Z
, et al
.
Long noncoding RNA Tug1 regulates mitochondrial bioenergetics in diabetic nephropathy
.
J Clin Invest
2016
;
126
:
4205
4218
[PubMed]
11.
Tang
PM
,
Zhang
YY
,
Mak
TS
,
Tang
PC
,
Huang
XR
,
Lan
HY
.
Transforming growth factor-β signalling in renal fibrosis: from Smads to non-coding RNAs
.
J Physiol
2018
;
596
:
3493
3503
[PubMed]
12.
Zhou
Q
,
Chung
AC
,
Huang
XR
,
Dong
Y
,
Yu
X
,
Lan
HY
.
Identification of novel long noncoding RNAs associated with TGF-β/Smad3-mediated renal inflammation and fibrosis by RNA sequencing
.
Am J Pathol
2014
;
184
:
409
417
[PubMed]
13.
Feng
M
,
Tang
PMK
,
Huang
XR
, et al
.
TGF-β mediates renal fibrosis via the smad3-Erbb4-IR long noncoding RNA Axis
.
Mol Ther
2018
;
26
:
148
161
[PubMed]
14.
Sun
SF
,
Tang
PMK
,
Feng
M
, et al
.
Novel lncRNA Erbb4-IR promotes diabetic kidney injury in db/db mice by targeting miR-29b
.
Diabetes
2018
;
67
:
731
744
[PubMed]
15.
Qin
W
,
Chung
ACK
,
Huang
XR
, et al
.
TGF-β/Smad3 signaling promotes renal fibrosis by inhibiting miR-29
.
J Am Soc Nephrol
2011
;
22
:
1462
1474
[PubMed]
16.
Zhong
X
,
Chung
ACK
,
Chen
HY
,
Meng
XM
,
Lan
HY
.
Smad3-mediated upregulation of miR-21 promotes renal fibrosis
.
J Am Soc Nephrol
2011
;
22
:
1668
1681
[PubMed]
17.
Li
R
,
Chung
ACK
,
Dong
Y
,
Yang
W
,
Zhong
X
,
Lan
HY
.
The microRNA miR-433 promotes renal fibrosis by amplifying the TGF-β/Smad3-Azin1 pathway
.
Kidney Int
2013
;
84
:
1129
1144
[PubMed]
18.
Zhou
Q
,
Huang
XR
,
Yu
J
,
Yu
X
,
Lan
HY
.
Long noncoding RNA Arid2-IR is a novel therapeutic target for renal inflammation
.
Mol Ther
2015
;
23
:
1034
1043
[PubMed]
19.
Kong
L
,
Zhang
Y
,
Ye
ZQ
, et al
.
CPC: assess the protein-coding potential of transcripts using sequence features and support vector machine
.
Nucleic Acids Res
2007
;
35
:
W345
W349
[PubMed]
20.
Wang
L
,
Park
HJ
,
Dasari
S
,
Wang
S
,
Kocher
JP
,
Li
W
.
CPAT: Coding-Potential Assessment Tool using an alignment-free logistic regression model
.
Nucleic Acids Res
2013
;
41
:e74
[PubMed]
21.
Chen
X
,
Song
E
.
Turning foes to friends: targeting cancer-associated fibroblasts
.
Nat Rev Drug Discov
2019
;
18
:
99
115
[PubMed]
22.
Zimmerman
SG
,
Peters
NC
,
Altaras
AE
,
Berg
CA
.
Optimized RNA ISH, RNA FISH and protein-RNA double labeling (IF/FISH) in Drosophila ovaries
.
Nat Protoc
2013
;
8
:
2158
2179
[PubMed]
23.
Piek
E
,
Ju
WJ
,
Heyer
J
, et al
.
Functional characterization of transforming growth factor beta signaling in Smad2- and Smad3-deficient fibroblasts
.
J Biol Chem
2001
;
276
:
19945
19953
[PubMed]
24.
Duan
WJ
,
Yu
X
,
Huang
XR
,
Yu
JW
,
Lan
HY
.
Opposing roles for Smad2 and Smad3 in peritoneal fibrosis in vivo and in vitro
.
Am J Pathol
2014
;
184
:
2275
2284
[PubMed]
25.
Tang
PMK
,
Zhou
S
,
Meng
XM
, et al
.
Smad3 promotes cancer progression by inhibiting E4BP4-mediated NK cell development
.
Nat Commun
2017
;
8
:
14677
[PubMed]
26.
You
YK
,
Huang
XR
,
Chen
HY
,
Lyu
XF
,
Liu
HF
,
Lan
HY
.
C-reactive protein promotes diabetic kidney disease in db/db mice via the CD32b-smad3-mTOR signaling pathway
.
Sci Rep
2016
;
6
:
26740
[PubMed]
27.
Lv
LL
,
Tang
PM
,
Li
CJ
, et al
.
The pattern recognition receptor, Mincle, is essential for maintaining the M1 macrophage phenotype in acute renal inflammation
.
Kidney Int
2017
;
91
:
587
602
[PubMed]
28.
Wang
QM
,
Tang
PM
,
Lian
GY
, et al
.
Enhanced cancer immunotherapy with smad3-silenced NK-92 cells
.
Cancer Immunol Res
2018
;
6
:
965
977
[PubMed]
29.
Tang
PM
,
Zhou
S
,
Li
CJ
, et al
.
The proto-oncogene tyrosine protein kinase Src is essential for macrophage-myofibroblast transition during renal scarring
.
Kidney Int
2018
;
93
:
173
187
[PubMed]
30.
Chung
ACK
,
Zhang
H
,
Kong
YZ
, et al
.
Advanced glycation end-products induce tubular CTGF via TGF-beta-independent Smad3 signaling
.
J Am Soc Nephrol
2010
;
21
:
249
260
[PubMed]
31.
Li
JH
,
Huang
XR
,
Zhu
HJ
, et al
.
Advanced glycation end products activate Smad signaling via TGF-beta-dependent and independent mechanisms: implications for diabetic renal and vascular disease
.
FASEB J
2004
;
18
:
176
178
[PubMed]
32.
Barutta
F
,
Bruno
G
,
Grimaldi
S
,
Gruden
G
.
Inflammation in diabetic nephropathy: moving toward clinical biomarkers and targets for treatment
.
Endocrine
2015
;
48
:
730
742
[PubMed]
33.
Duncan
BB
,
Schmidt
MI
,
Pankow
JS
, et al.;
Atherosclerosis Risk in Communities Study
.
Low-grade systemic inflammation and the development of type 2 diabetes: the atherosclerosis risk in communities study
.
Diabetes
2003
;
52
:
1799
1805
[PubMed]
34.
Jagannathan-Bogdan
M
,
McDonnell
ME
,
Shin
H
, et al
.
Elevated proinflammatory cytokine production by a skewed T cell compartment requires monocytes and promotes inflammation in type 2 diabetes
.
J Immunol
2011
;
186
:
1162
1172
[PubMed]
35.
Tang
PM
,
Nikolic-Paterson
DJ
,
Lan
HY
.
Macrophages: versatile players in renal inflammation and fibrosis
.
Nat Rev Nephrol
2019
;
15
:
144
158
[PubMed]
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 http://www.diabetesjournals.org/content/license.

Supplementary data