key: cord-0001949-mqk0sx5n authors: Lo, Chao-Sheng; Shi, Yixuan; Chang, Shiao-Ying; Abdo, Shaaban; Chenier, Isabelle; Filep, Janos G.; Ingelfinger, Julie R.; Zhang, Shao-Ling; Chan, John S. D. title: Overexpression of heterogeneous nuclear ribonucleoprotein F stimulates renal Ace-2 gene expression and prevents TGF-β1-induced kidney injury in a mouse model of diabetes date: 2015-08-01 journal: Diabetologia DOI: 10.1007/s00125-015-3700-y sha: 8352a3f13888fc4958f6b96a7dc06aed2724f52e doc_id: 1949 cord_uid: mqk0sx5n AIMS/HYPOTHESIS: We investigated whether heterogeneous nuclear ribonucleoprotein F (hnRNP F) stimulates renal ACE-2 expression and prevents TGF-β1 signalling, TGF-β1 inhibition of Ace-2 gene expression and induction of tubulo-fibrosis in an Akita mouse model of type 1 diabetes. METHODS: Adult male Akita transgenic (Tg) mice overexpressing specifically hnRNP F in their renal proximal tubular cells (RPTCs) were studied. Non-Akita littermates and Akita mice served as controls. Immortalised rat RPTCs stably transfected with plasmid containing either rat Hnrnpf cDNA or rat Ace-2 gene promoter were also studied. RESULTS: Overexpression of hnRNP F attenuated systemic hypertension, glomerular filtration rate, albumin/creatinine ratio, urinary angiotensinogen (AGT) and angiotensin (Ang) II levels, renal fibrosis and profibrotic gene (Agt, Tgf-β1, TGF-β receptor II [Tgf-βrII]) expression, stimulated anti-profibrotic gene (Ace-2 and Ang 1–7 receptor [MasR]) expression, and normalised urinary Ang 1–7 level in Akita Hnrnpf-Tg mice as compared with Akita mice. In vitro, hnRNP F overexpression stimulated Ace-2 gene promoter activity, mRNA and protein expression, and attenuated Agt, Tgf-β1 and Tgf-βrII gene expression. Furthermore, hnRNP F overexpression prevented TGF-β1 signalling and TGF-β1 inhibition of Ace-2 gene expression. CONCLUSIONS/INTERPRETATION: These data demonstrate that hnRNP F stimulates Ace-2 gene transcription, prevents TGF-β1 inhibition of Ace-2 gene transcription and induction of kidney injury in diabetes. HnRNP F may be a potential target for treating hypertension and renal fibrosis in diabetes. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1007/s00125-015-3700-y) contains peer-reviewed but unedited supplementary material, which is available to authorised users. Diabetic nephropathy (DN), a leading cause of endstage renal disease (ESRD), accounts for ∼50% of all ESRD cases [1, 2] . While glomerulopathy is a hallmark of early renal injury in DN [3] , tubulointerstitial fibrosis and tubular atrophy are major features of late-stage DN and are closely associated with loss of renal function [4] [5] [6] [7] . The mechanisms underlying tubulointerstitial fibrosis, however, are incompletely understood. TGF-β1 is considered to be the most potent inducer of fibrogenesis [8] . Indeed, patients and animal models with type 1 or 2 diabetes have significantly elevated serum and urinary TGF-β1 levels [9] [10] [11] as well as heightened TGF-β1 mRNA and protein expression in glomeruli and the tubulointerstitium [12] [13] [14] [15] [16] . We previously reported that high glucose milieu enhances expression of angiotensinogen (AGT, the sole precursor of all angiotensins) through generation of reactive oxygen species (ROS) in cultured rat renal proximal tubular cells (RPTCs) [17, 18] . Rat AGT overexpression in RPTCs leads to hypertension, albuminuria and RPTC hypertrophy, and enhances TGF-β1 expression in diabetic AGT-transgenic (Tg) mice [19, 20] . Conversely, RPTC-selective overexpression of catalase or pharmacological blockade of the renin-angiotensin system (RAS) attenuates hypertension, ROS generation, kidney injury and normalised RPTC ACE-2 expression in mouse models of diabetes [21] [22] [23] [24] . Taken together, these observations indicate that oxidative stress-induced upregulation of AGT expression and downregulation of ACE-2 expression in RPTCs, resulting in higher angiotensin (Ang)II/Ang 1-7 ratio, may be key determinants of development of hypertension and nephropathy in diabetes. We reported that insulin inhibits high glucose stimulation of rat renal Agt gene expression via two nuclear proteins-heterogeneous nuclear ribonucleoproteins F and K (hnRNP F, hnRNP K)-that interact with the insulin-responsive element (IRE) in the Agt gene promoter [25] [26] [27] [28] , and that hnRNP F overexpression in RPTCs inhibits Agt gene expression and kidney hypertrophy in Akita Hnrnpf-Tg mice [29] . Here, we report that overexpression of hnRNP F stimulates Ace-2 gene transcription and suppresses profibrotic gene (Tgf-β1, Tgf-βrII) expression in RPTCs of Akita Hnrnpf-Tg mice. We have confirmed these changes by in vitro studies in rat RPTCs. We also show that hnRNP F overexpression prevents TGF-β1 signalling and inhibition of Ace-2 gene expression in RPTCs. Finally, we identified the putative DNA response elements (REs) in the Ace-2 gene promoter that are responsive to hnRNP F and TGF-β1. Chemicals and constructs Active human recombinant TGF-β1 was obtained from R&D Systems (Minneapolis, MN, USA). SB431542 (a TGF-β receptor I [RI] inhibitor) and other chemicals were purchased from Sigma-Aldrich (Oakville, ON, Canada). The antibodies used in the present study are listed in electronic supplementary material (ESM) Table 1. The pKAP2 plasmid containing the kidney-specific androgen-regulated protein (KAP) promoter was a gift from C. D. Sigmund (University of Iowa, Iowa City, IA, USA) [30] . Full-length rat Hnrnpf cDNA fused with HA tag (encoding amino acid residues 98-106 [YPYDVPDYA] of human influenza virus hemagglutinin) was inserted into pKAP2 plasmid at the NotI site at both 5′ and 3′ termini [25, 29] . pGL4.20 vector containing Luciferase reporter was obtained from Promega (Sunnyvale, CA, USA). Rat Ace-2 gene promoter (N-1,091/+83) was cloned from rat genomic DNA with specific primers (ESM Table 2 ), as described by Milsted et al [31] and then inserted into pGL4.20 plasmid at HindIII and KpnI restriction sites. Scrambled Silencer Negative Control no. 1 small interfering RNA (siRNA) and Hnrnpf siRNA were bought from Ambion (Austin, TX, USA). QuickChange II Site-Directed Mutagenesis Kit and LightShift Chemiluminescent electrophoretic mobility shift assay (EMSA) Kit were procured from Agilent Technologies (Santa Clara, CA, USA) and Thermo Scientific (Life Technologies, Burlington, ON, Canada), respectively. The primer biotinlabelling kit was purchased from Integrated DNATechnologies (Coralville, IA, USA). Physiological studies Adult male heterozygous Akita mice (Mus musculus) with a mutated Ins2 gene (C57BL6-Ins2 Akita /J) were purchased from Jackson Laboratories (Bar Harbor, ME, USA: http://jaxmice.jax.org). Akita Tg mice (C57Bl/6 background) overexpressing rat hnRNP F-HA in RPTCs (line 937) were created in our laboratory (by J. S. D. Chan) [29] . Male adult non-Tg and non-Akita littermates served as wild-type (WT) controls, and were tested along with Hnrnpf-Tg, Akita and Akita Hnrnpf-Tg mice. All animals were housed individually in metabolic cages for 24 h before euthanasia at age 20 weeks. All animals were fed standard mouse chow and water ad libitum. Animal care and procedures were approved by the CRCHUM Animal Care Committee and followed the Principles of Laboratory Animal Care (NIH publication no. 85-23, revised 1985: http:// grants1.nih.gov/grants/olaw/references/phspol.htm). Blood glucose levels, following 4-5 h fasting, were determined with an Accu-Chek Performa System (Roche Diagnostics, Laval, QC, Canada). Body weight (BW) was recorded. Urine was collected and assayed for albumin/creatinine ratio (ACR) by enzyme-linked immunosorbent assays (Albuwell and Creatinine Companion, Exocell, Philadelphia, PA, USA). GFR was measured as described by Qi et al [32] as recommended by the Animal Models of Diabetic Complications Consortium (www.diacomp.org) with fluorescein isothiocyanate inulin [23, 28, 33] . Kidneys were removed immediately after GFR measurement, decapsulated and weighed. The left kidneys were processed for histology and immunostaining, and right renal cortices were harvested for renal proximal tubules (RPTs) isolation by Percoll gradient centrifugation [23, 24, 28, 29] . Aliquots of freshly isolated RPTs from individual mice were immediately processed for total RNA and protein isolation. Immunohistochemical staining Immunohistochemical staining was performed by the standard avidin-biotin-peroxidase complex method in four to five sections (4 μm thick) per kidney and three mouse kidneys per group (ABC Staining System; Santa Cruz Biotechnology [Santa Cruz, CA, USA]) [23, 24, 28, 29] . Staining was analysed under light microscopy by two independent, blinded observers. The collected images were assessed by National Institutes of Health Image J software (http://rsb.info.nih.gov/ij/) [23, 24, 28, 29] . Urinary AGT, Ang II and Ang 1-7 measurement Mouse urinary AGT, Ang II and Ang 1-7 levels were analysed by ELISA (Immuno-Biological Laboratories, IBL America, Minneapolis, MN, USA) and normalised by urinary creatinine levels as described [23, 24, 28, 29, 34] . Cell culture Immortalised rat RPTCs (passages 12-18) [35] were cultured in 5 mmol/l D-glucose DMEM containing 5% FBS until they reached 60-70% confluence. The media were then changed to serum-free DMEM, ensuring that endogenously secreted TGF-β1 would not interfere in the assay. After 45 min preincubation, active human recombinant TGF-β1 [36] (0 to 10 ng/ml) was added (considered as time 0 h) and incubated for various time periods up to 24 h. In separate experiments, RPTCs were incubated for 24 h in serum-free medium in the presence or absence of TGF-β1± various concentrations of SB431542. Real-time quantitative PCR Hnrnpf, Ace, Ace-2, MasR, Tgf-β1, Tgf-βrI, Tgf-βrII, collagen type IV, collagen type I, fibronectin 1 and β-actin mRNA expression levels in RPTs were quantified by real-time quantitative PCR (RT-qPCR) with forward and reverse primers (ESM Table 2 ) [23, 24, 28, 29] . Western blotting Western blotting (WB) was performed as described previously [23, 24, 28, 29] . The relative densities of hnRNP F, ACE, ACE-2, Ang 1-7 receptor (MasR), TGF-β1, TGF-β RI, TGF-β RII, fibronectin 1, p-Smad2/3, Smad2/3 Akita compared with WT mice and Hnrnpf-Tg mice. Overexpression of hnRNP F had no effect on blood glucose levels in Akita Hnrnpf-Tg mice. Systolic BP (SBP), kidney weight (KW)/BW and KW/tibial length (TL) ratios, GFR and ACR were all elevated in Akita mice, compared with both WT controls and Hnrnpf-Tg mice. HnRNP F overexpression in RPTCs markedly attenuated these changes in diabetic Akita Hnrnpf-Tg mice. Furthermore, Akita mice exhibited elevated urinary AGT and Ang II levels, parallel with decreased Ang 1-7 levels, compared with WT mice. HnRNP F overexpression partially reduced urinary AGT and Ang II levels, whereas it completely normalised urinary Ang 1-7 levels-a novel finding. Effect of hnRNP F overexpression on AGT, ACE, ACE-2 and MasR expression in Akita Hnrnpf-Tg mouse kidneys Immunostaining revealed that HnRNP F (Fig. 1a) was overexpressed in RPTCs of Hnrnpf-Tg and Akita Hnrnpf-Tg mice compared with WT and Akita mice, respectively. ACE-2 ( Fig. 1b) and MasR (Fig. 1c ) expression was decreased in Akita mice compared with WT controls and normalised in Akita Hnrnpf-Tg mice. RPTC ACE (Fig. 1d ) expression did not differ between WT and Hnrnpf-Tg mice, whereas ACE expression was significantly higher in Akita mice than in WT controls and was not normalised in Akita Hnrnpf-Tg mice. WB and RT-qPCR for hnRNP F, ACE-2, MasR and ACE protein and their mRNA levels ( Fig. 1e -l, respectively) confirmed these observations. Effect of hnRNP F overexpression on TGF-β1, TGF-β RII and TGF-β RI expression in Akita Hnrnpf-Tg mouse kidneys Immunostaining of TGF-β1 (Fig. 2a) and TGF-β RII (Fig. 2b) , WB of TGF-β1 (Fig. 2d) and TGF-β RII expression Values are means+SEM corrected to β-actin, n=6. *p<0.05; **p<0.01 (Fig. 2e) , and RT-qPCR of Tgf-β1 (Fig. 2g) and Tgf-βrII (Fig. 2h ) mRNA expression showed significantly higher TGF-β1 and TGF-β RII expression in RPTCs of Akita mice than in WT controls and Hnrnpf-Tg mice, and they were attenuated in Akita Hnrnpf-Tg mice. In contrast, TGF-β RI expression was similar in all groups studied (Fig. 2c,f,i) . HnRNP F overexpression suppresses renal fibrosis in Akita Hnrnpf-Tg mice Akita mice developed renal structural damage compared with WT and Hnrnpf-Tg mice (ESM Fig. 1a , PAS staining), including tubular luminal dilatation with accumulation of cell debris, increased extracellular matrix proteins in glomeruli and tubules, and proximal tubule cell atrophy. HnRNP F overexpression markedly reversed but never completely resolved these abnormalities in Akita mice. We detected significant increases in Masson's trichrome staining (Fig. 3a) and immunostaining for collagen type IV (Fig. 3b) , fibronectin 1 expression (Fig. 3c ) and collagen type I (Fig. 3d) in glomerulotubular areas in Akita mice compared with WT controls and Hnrnpf-Tg mice. These changes were reduced in Akita Hnrnpf-Tg mice. Quantification of Masson's trichrome-stained (ESM Fig. 1b) , immunostaining of collagen IV (Fig. 3e) , fibronectin 1 (Fig. 3f) and collagen I (Fig. 3g) , and RT-qPCR quantification of mRNA levels (Fig. 3h-j) confirmed their expression. HnRNP F overexpression enhances Ace-2 and suppresses Agt, Tgf-β1 and Tgf-βrII gene expression and protein levels in rat RPTCs in vitro RPTCs stably transfected with pcDNA 3.1/Hnrnpf (RPTC-pcDNA 3.1/Hnrnpf) exhibited considerably higher levels of hnRNP F (Fig. 4a,b) , lower amounts of AGT (Fig. 4a,c) and a higher amount of ACE-2 (Fig. 4a,d) than non-transfected RPTCs or RPTCs stably transfected with pcDNA 3.1 (RPTC-pcDNA 3.1). In contrast, TGF-β1 and TGF-β RII protein levels were significantly decreased in RPTC-pcDNA 3.1/Hnrnpf compared with non-transfected RPTCs or RPTC-pcDNA 3.1 (p<0.01) (Fig. 4e,f,g, respectively) . TGF-β RI protein level was similar in non-transfected RPTCs, RPTC-pcDNA 3.1 or RPTC-pcDNA 3.1/Hnrnpf (Fig. 4h) . RT-qPCR of Hnrnpf, Agt, Ace-2, Tgf-β1, Tgf-βrII and Tgf-βrI mRNA levels confirmed these findings (ESM Fig. 2a-f ). TGF-β1 signalling and inhibition of Ace-2 gene expression in rat RPTCs TGF-β1 inhibited rat Ace-2 gene promoter activity (Fig. 5a) , rat Ace-2 mRNA expression (Fig. 5b) and rat ACE-2 protein level (Fig. 5c) in a concentration-dependent manner, which was reversed by SB431542 (a TGF-β RI inhibitor) (Fig. 5d-f, respectively) . Furthermore, TGF-β1 stimulated Smad 2/3 phosphorylation in a concentration-and timedependent manner (Fig. 5g) and reversed by SB431542 (Fig. 5h) . These data demonstrate that TGF-β1 inhibition of Ace-2 gene transcription is mediated, at least in part, via Smad2/3 signalling. HnRNP F overexpression prevents TGF-β signalling, and TGF-β inhibition of Ace-2 and induction of fibrotic gene expression in RPTCs TGF-β1 had no detectable effect on hnRNP F protein levels (Fig. 6a,b) . Intriguingly, hnRNP F overexpression prevented TGF-β1 stimulation of Smad 2/3 phosphorylation (Fig. 6a,c) , TGF-β RII expression (Fig. 6a , d) and fibronectin 1 expression (Fig. 6a,e) . HnRNP F overexpression also prevented TGF-β1-induced downregulation of MasR (Fig. 6a ,f) content in RPTCs. Addition of TGF-β1 did not affect TGF-β RI expression in RPTCs (Fig. 6a,g) . Furthermore, overexpression of hnRNP F prevented the inhibitory effect of TGF-β1 on ACE-2 protein (Fig. 6a,h) and Ace-2 mRNA (Fig. 6i ) expression in RPTC-pcDNA 3.1/ Hnrnpf. Localisation of Hnrnpf-and TGF-β1 (or SMAD)-RE in rat Ace-2 gene promoter To localise the putative DNA-RE(s) that mediate(s) the action of hnRNP F or TGF-β1 on Ace-2 gene promoter activity, plasmids containing various lengths of the rat Ace-2 gene promoter were transiently transfected into RPTC-pcDNA 3.1 or RPTC-pcDNA 3.1/ Hnrnpf. The activity of pGL4.20-Ace-2 promoter (N-1,091/+ 83) and pGL 4.20-Ace-2 promoter (N-499/+83) exhibited respective fivefold and 12-fold increase as compared with the control plasmid, pGL 4.20 in RPTC-pcDNA 3.1 (Fig. 7a) . Further deletion of nucleotides N-499 to N-241 (pGL 4.20-Ace-2 promoter [N-240/+83]) significantly reduced the rat Ace-2 promoter activity. Moreover, the activity of pGL4.20-Ace-2 promoter (N-1,091/+83) and pGL4.20-Ace-2 promoter (N-499/+83) was further increased by 1.5-2.0-fold, whereas the activity of pGL4.20-Ace-2 promoter (N-240/+83) did not increase in RPTC-pcDNA 3.1/Hnrnpf as compared with RPTC-pcDNA 3.1 (Fig. 7a) . Interestingly, addition of TGF-β1 inhibited the promoter activity of pGL 4.20-Ace-2 promoter (N-1,091/+83) and did not affect the activity of pGL 4.20-Ace-2 promoter (N-499/+83) and pGL 4.20-Ace-2 promoter (N-240/+83) in RPTC-pcDNA 3.1 (Fig. 7b) . However, TGF-β1 had no inhibitory effect on the promoter activity of these constructs in RPTC-pcDNA 3.1/Hnrnpf (Fig. 7c) . In contrast, transfection of Hnrnpf siRNA significantly inhibited the promoter activity of pGL 4.20-Ace-2 promoter (N-1,091/+83) and pGL 4.20-Ace-2 promoter (N-499/+83) without affecting the activity of pGL 4.20-Ace-2 promoter (N-240/+83) in RPTC-pcDNA 3.1 (Fig. 7d) . Deletion of the nucleotides N-401 to N-393 (5′-ggggagagg-3′) in the Ace-2 gene promoter markedly attenuated the promoter activity of pGL 4.20-Ace-2 promoter (N-1,091/+83) and pGL 4.20-Ace-2 promoter (N-499/+83) in RPTC-pcDNA 3.1/Hnrnpf (Fig. 7e) . Interestingly, deletion of the putative proximal SMAD-RE (nucleotides N-511 to N-504 [5′-cagagaca-3′]) or distal putative SMAD-RE2 (nucleotides N-789 to N-784 [5′-gagaca-3′]) in the Ace-2 gene promoter partially attenuated whereas deletion of both REs (nucleotides N-511 to N-504 and nucleotides N-789 to N-784) completely abolished the inhibitory action of TGF-β1 on pGL 4.20-Ace-2 promoter (N-1,091/+83) activity in RPTC-pcDNA 3.1 (Fig. 7f) . Furthermore, EMSA showed that the double strand DNA fragments, nucleotides N-405 to N-387 (putative Hnrnpf-RE), nucleotides N-518 to N-497 (putative proximal SMAD-RE1) and nucleotides N-797 to N-776 (putative distal SMAD-RE2) bind to the nuclear proteins from RPTCs and they could be displaced by the respective WT DNA fragments, but not by mutated DNA fragments (Fig. 7g,h, respectively) . Importantly, addition of anti-hnRNP F and anti-Smad 2/3 antibody induced a supershift of the respective Hnrnpf-RE and SMAD-REs with the nuclear proteins, respectively (Fig. 7g,h) . The present report identifies a novel mechanism by which hnRNP F prevents hypertension and kidney injury in diabetic Akita mice, i.e. hnRNP F stimulation of renal Ace-2 gene transcription and mitigation of the inhibitory effect of TGF-β1 on Ace-2 gene transcription. We reported previously that overexpression of hnRNP F prevents systemic hypertension, and inhibits renal Agt gene expression and RPTC hypertrophy in diabetic Akita Hnrnpf-Tg mice [29] . The present paper provides new in vivo and in vitro evidence that hnRNP F stimulates Ace-2 gene transcription via binding to the DNA-RE of the Ace-2 gene promoter, which is critical for the formation of renal Ang 1-7 and subsequent expression of its antihypertensive and renoprotective actions in Akita mice [37] . HnRNP F, a member of the pre-mRNA-binding protein family [38] regulates gene expression at both the transcriptional and post-transcriptional levels. Indeed, hnRNP F engages in alternative splicing of various genes [39] [40] [41] and associates with TATA-binding protein, RNA polymerase II, nuclear cap-binding protein complex and various transcriptional factors. [42, 43] The Akita mouse is an autosomal-dominant model of spontaneous type 1 diabetes in which the Ins2 gene is mutated. Akita mice develop hyperglycaemia and systemic hypertension, leading to cardiac hypertrophy, left ventricular diastolic dysfunction, glomerulosclerosis and enhanced oxidative stress in RPTs, closely resembling those observed in patients with type 1 diabetes [44, 45] . Our study provides evidence for a novel mechanism for hnRNP F lowering of SBP: inhibition of intrarenal Agt gene expression and RAS activation, concomitant with upregulation of the ACE-2/Ang 1-7/MasR axis. Indeed, our results show that hnRNP F overexpression inhibited renal AGT and Agt mRNA expression (ESM Fig. 1 c-e), lowered urinary AGT and Ang II levels and normalised urinary Ang 1-7 levels. We consistently observed decreased renal ACE-2 expression in Akita mice as previously reported [23, 24] . Decreased ACE-2 expression also has been reported in male streptozotocin (STZ)-induced diabetic mice [46] , STZinduced diabetic rats [47, 48] and human type 2 diabetic kidneys [49, 50] . The precise mechanism by which hnRNP F overexpression leads to upregulation of renal Ace-2 and MasR gene expression in diabetes remains unclear. One possibility is that hnRNP F binds to putative Hnrnpf-RE(s) in the Ace-2 and MasR gene promoters, subsequently enhancing Ace-2 and MasR gene transcription. This possibility is supported by our findings that hnRNP F considerably augments the activity of an Ace-2 gene promoter and that the Hnrnpf siRNA and deletion of the putative Hnrnpf-RE markedly reduced the rat Ace-2 gene promoter activity in RPTCs. Furthermore, the biotinylated-labelled Hnrnpf-RE specifically bound to RPTC nuclear proteins and the addition of anti-hnRNP F antibody yielded a supershift of biotinylated-labelled Hnrnpf-RE binding with nuclear proteins in EMSA. These data demonstrate that hnRNP F binds to the putative Hnrnpf-RE and stimulates Ace-2 gene transcription. Of note, hnRNP F is not specific for ---+ + + + + + + + + + + -BSA + + + + + + + + + Ace-2 gene expression but also affects the expression of Agt [25] and other genes [51, 52] . Currently, little is known about the mechanisms by which TGF-β1 downregulates renal Ace-2 gene expression in diabetes. Chou et al [53] reported that SB431542 inhibited high glucose and TGF-β1 inhibition of Ace-2 mRNA expression in cultured NRK-52 cells. Our findings confirm these observations. Our present studies also demonstrate that TGF-β1 inhibits the activity of pGL 4.20-rat Ace-2 promoter (N-1, 091/+83) and that deletion of putative SMAD-REs in the Ace-2 gene promoter mitigates the inhibitory effect of TGF-β1 on the Ace-2 gene promoter activity. Furthermore, biotinylated-labelled SMAD-REs bound to RPTC nuclear proteins and the addition of anti-Smad2/3 antibody yielded a supershift of labelled DNA with nuclear proteins. These data demonstrate that the inhibitory effect of TGF-β1 on Ace-2 gene transcription is mediated, at least in part, via the SMAD-REs in the Ace-2 gene promoter. Intriguingly, hnRNP F overexpression prevented TGF-β1 signalling on Smad2/3 phosphorylation and on TGF-β1 inhibition of Ace-2 gene promoter activity in RPTCs. At present, the underlying molecular mechanism of how hnRNP F prevents TGF-β1 inhibition of Ace-2 gene transcription is not yet defined. One possibility might be that hnRNP F directly inhibits Tgf-β1rII gene expression as shown in our studies. The second possibility is that hnRNP F might interfere or prevent the interaction of Smad2/3 with other transcriptional factor(s) to inhibit Ace-2 gene transcription. Clearly, more studies are needed to define the molecular interaction of hnRNP F with Smad2/3 on Ace-2 gene transcription. In summary, the present study suggests a major role for hnRNP F in attenuating systemic hypertension and renal fibrosis in experimental diabetes and possibly in diabetic human kidneys. Our observations raise the possibility that selective targeting of this antihypertensive and anti-fibrotic protein may represent a novel approach for preventing or reversing the pathological manifestations of DN, particularly tubular fibrosis. Identification of Hnrnpf-RE and SMAD-RE in the Ace-2 gene promoter. (a) Luciferase activity of the plasmid containing various lengths of Ace-2 gene promoter in RPTC-pcDNA 3.1 (white bars) and in RPTC-pcDNA 3.1/Hnrnpf (black bars) ng/ml hTGF-β1); and (c) in RPTC-pcDNA3.1/Hnrnpf±TGF-β1 (white bars, without hTGF-β1; black bars, with 2 ng/ml hTGF-β1); (d) in RPTC-pcDNA 3.1±Hnrnpf siRNA (white bars, treated with 50 nmol/l scrambled siRNA; black bars, treated with 50 nmol/l Hnrnpf siRNA), cultured in normal glucose media for 24 h. (e) Promoter activity of the Ace-2 gene ±Hnrnpf black bars) or (f)±SMAD-REs in RPTC-pcDNA 3.1 in the absence or presence of TGF-β1 (white bars, without hTGF-β1; black bars, with 2 ng/ml hTGF-β1). Values are mean+SEM, n=6. The experiments were repeated twice REs (h) with RPTC nuclear proteins±excess unlabelled WT Hnrnpf-RE or mutated Hnrnpf-REs (M1 to M4 are mutants of Hnrnpf-RE with nucleotides mutated or deleted in the binding motif as shown in ESM Table 2) or WT SMAD-RE or mutant SMAD-REs (SMAD-RE1 [M1 and M2] and SMAD-RE2 [M1 and M2] are mutants of respective SMAD-RE1 and SMAD-RE2 with nucleotides mutated in the binding motif as shown in ESM Table 2). Rabbit IgG or rabbit anti-hnRNP F or anti-Smad2/3 antiserum was added to the reaction mixture and incubated for 30 min on ice before incubation with the biotinylated probe. Results are representative of three independent experiments Endogenous angiotensin concentrations in specific intrarenal fluid compartments of the rat The early natural history of nephropathy in type 1 diabetes: II. Early renal structural changes in type 1 diabetes Mechanisms of tubulointerstitial injury in the kidney: final common pathways to end-stage renal failure The tubulointerstitium in progressive diabetic kidney disease: more than an aftermath of glomerular injury? Significance of tubulointerstitial changes in the renal cortex for the excretory function and concentration ability of the kidney: a morphometric contribution Tubulointerstitial damage leads to atubular glomeruli: significance and possible role in progression Transforming growth factorbeta regulates tubular epithelial-myofibroblast transdifferentiation in vitro Accurate measurement and clinical significance of urinary transforming growth factor-beta1 Utility of serum and urinary transforming growth factor-beta levels as markers of diabetic nephropathy Increased excretion of urinary transforming growth factor beta 1 in patients with diabetic nephropathy Evidence that TGF-beta should be a therapeutic target in diabetic nephropathy Increased renal production of transforming growth factor-beta1 in patients with type II diabetes Expression of transforming growth factor beta is elevated in human and experimental diabetic nephropathy The renal expression of transforming growth factor-beta isoforms and their receptors in acute and chronic experimental diabetes in rats Increased glomerular and tubular expression of transforming growth factor-beta1, its type II receptor, and activation of the Smad signaling pathway in the db/db mouse High glucose stimulates angiotensinogen gene expression via reactive oxygen species generation in rat kidney proximal tubular cells High glucose stimulates angiotensinogen gene expression and cell hypertrophy via activation of the hexosamine biosynthesis pathway in rat kidney proximal tubular cells RAS blockade decreases blood pressure and proteinuria in transgenic mice overexpressing rat angiotensinogen gene in the kidney Overexpression of angiotensinogen increases tubular apoptosis in diabetes Catalase overexpression attenuates angiotensinogen expression and apoptosis in diabetic mice Attenuation of interstitial fibrosis and tubular apoptosis in db/db transgenic mice overexpressing catalase in renal proximal tubular cells Overexpression of catalase prevents hypertension and tubulointerstitial fibrosis and normalization of renal angiotensin-converting enzyme-2 expression in Akita mice Dual RAS blockade normalizes angiotensin-converting enzyme-2 expression and prevents hypertension and tubular apoptosis in Akita angiotensinogen-transgenic mice Heterogenous nuclear ribonucleoprotein F modulates angiotensinogen gene expression in rat kidney proximal tubular cells Characterization of a putative insulin-responsive element and its binding protein(s) in rat angiotensinogen gene promoter: regulation by glucose and insulin Heterogeneous nuclear ribonucleoprotein K modulates angiotensinogen gene expression in kidney cells Heterogeneous nuclear ribonucleoproteins F and K mediate insulin inhibition of renal angiotensinogen gene expression and prevention of hypertension and kidney injury in diabetic mice Heterogeneous nuclear ribonucleoprotein F suppresses angiotensinogen gene expression and attenuates hypertension and kidney injury in diabetic mice The kidney androgenregulated protein promoter confers renal proximal tubule cell-specific and highly androgen-responsive expression on the human angiotensinogen gene in transgenic mice Regulation of multiple renin-angiotensin system genes by Sry Characterization of susceptibility of inbred mouse strains to diabetic nephropathy Angiotensin II type II receptor deficiency accelerates the development of nephropathy in type I diabetes via oxidative stress and ACE2 Catalase overexpression prevents hypertension and tubular apoptosis in angiotensinogen transgenic mice Rat proximal tubule cell line transformed with origin-defective SV40 DNA: autocrine ANG II feedback Transforming growth factor-beta 1 stimulates angiotensinogen gene expression in kidney proximal tubular cells Angiotensin-(1-7) prevents systemic hypertension, attenuates oxidative stress and tubulointerstitial fibrosis, and normalizes renal angiotensinconverting enzyme 2 and Mas receptor expression in diabetic mice Functional diversity of the hnRNPs: past, present and perspectives The generally expressed hnRNP F is involved in a neural-specific pre-mRNA splicing event Heterogeneous nuclear ribonucleoprotein F/H proteins modulate the alternative splicing of the apoptotic mediator Bcl-x Essential role for the interaction between hnRNP H/F and a G quadruplex in maintaining p53 pre-mRNA 3′-end processing and function during DNA damage Association of the rat heterogeneous nuclear RNA-ribonucleoprotein F with TATA-binding protein Interaction between the human nuclear cap-binding protein complex and hnRNP F A novel locus, Mody4, distal to D7Mit189 on chromosome 7 determines early-onset NIDDM in nonobese C57BL/6 (Akita) mutant mice Complications of IgA nephropathy in a non-insulin-dependent diabetes model, the Akita mouse ACE2 deficiency modifies renoprotection afforded by ACE inhibition in experimental diabetes Characterization of renal angiotensin-converting enzyme 2 in diabetic nephropathy Glomerular renin angiotensin system in streptozotocin diabetic and Zucker diabetic fatty rats Decreased glomerular and tubular expression of ACE2 in patients with type 2 diabetes and kidney disease Expression of ACE and ACE2 in individuals with diabetic kidney disease and healthy controls Proteomic identification of proteins associated with the osmoregulatory transcription factor TonEBP/OREBP: functional effects of Hsp90 and PARP-1 Global profiling of alternative splicing events and gene expression regulated by hnRNPH/F Interaction between TGF-beta and ACE2-Ang-(1-7)-Mas pathway in high glucosecultured NRK-52E cells Duality of interest The authors declare that there is no duality of interest associated with this manuscript.Contribution statement JSDC is the principal investigator and was responsible for the study conception and design. CSL drafted the manuscript and contributed to the discussion. CSL, YS, SYC, SA, IC and SLZ contributed to the in vivo and in vitro experiments and collection of data. JGF and JRI contributed to the Discussion and reviewed/edited the manuscript. All authors were involved in analysis and interpretation of data, and contributed to the critical revision of the manuscript. All authors provided final approval of the version to be published. JSDC is guarantor of this work and, as such, had full access to all study data, taking responsibility for data integrity and the accuracy of data analysis.Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http:// creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.