Abstract The pleiotropic benefits of statins in cardiovascular diseases that are independent of their lipid-lowering effects have been well documented, but the underlying mechanisms remain elusive. Here we show that simvastatin significantly improves human induced pluripotent stem cell-derived endothelial cell functions in both baseline and diabetic conditions by reducing chromatin accessibility at transcriptional enhanced associate domain elements and ultimately at endothelial-to-mesenchymal transition (EndMT)-regulating genes in a yes-associated protein (YAP)-dependent manner. Inhibition of geranylgeranyltransferase (GGTase) I, a mevalonate pathway intermediate, repressed YAP nuclear translocation and YAP activity via RhoA signaling antagonism. We further identified a previously undescribed SOX9 enhancer downstream of statin–YAP signaling that promotes the EndMT process. Thus, inhibition of any component of the GGTase–RhoA–YAP–SRY box transcription factor 9 (SOX9) signaling axis was shown to rescue EndMT-associated endothelial dysfunction both in vitro and in vivo, especially under diabetic conditions. Overall, our study reveals an epigenetic modulatory role for simvastatin in repressing EndMT to confer protection against endothelial dysfunction. __________________________________________________________________ Cardiovascular diseases (CVDs) are a leading cause of global mortality^[54]1. Statins, which are widely prescribed to reduce low-density lipoprotein cholesterol^[55]2, have pleiotropic activities that confer CVD protection beyond their lipid-lowering effect^[56]3–[57]5. While the precise mechanisms underlying these pleiotropic effects remain poorly understood, accumulating evidence suggests that epigenetic modifications play a key role in CVD initiation and progression^[58]6. For example, statins have been shown to protect against cancer and vascular diseases by remodeling chromatin accessibility through DNA methylation and histone acetylation^[59]7,[60]8. Thus, understanding of the role of statins in endothelial epigenetics could advance alternative therapeutic approaches to treat CVDs. Various human induced pluripotent stem cell (iPSC)-derived cardiovascular cells have been shown to accurately recapitulate drug responses in patients and to uncover pathogenic mechanisms of CVDs^[61]9. In this study, we integrated iPSC-derived endothelial cells (iPSC-ECs) and assay for transposase-accessible chromatin using sequencing (ATAC-seq) to show that simvastatin improved endothelial functions under baseline and diabetic or hyperglycemic (HG) conditions. It did so by decreasing chromatin accessibility at TEAD elements, such as the SOX9 enhancer region, and repressing the transcriptional activity of EndMT-regulating genes. We identified YAP as a direct regulator that enhanced chromatin accessibility at the SOX9 enhancer to upregulate EndMT genes. Simvastatin-regulated YAP activity was not affected by endothelial cell (EC) densities or mechanical perturbations. The inhibition of GGTase I, a key intermediate in the mevalonate pathway, was equally effective as simvastatin at improving endothelial functions by blocking YAP nuclear translocation and repressing EndMT in a RhoA-dependent manner. Genetic or pharmacological inhibition of any component of the GGTase–RhoA–YAP–SOX9 signaling axis effectively improved endothelial functions under both in vitro and in vivo diabetic conditions. In conclusion, our study identifies the mechanism underlying statin pleiotropy in endothelial protection via suppressing YAP-dependent chromatin remodeling at EndMT-regulating enhancer regions. The inhibition of YAP–SOX9 or their upstream regulators (that is, GGTase or RhoA) could represent alternative therapeutic strategies for treating endothelial dysfunction-associated CVDs, such as type 2 diabetes (T2D). Results Simvastatin improves endothelial functions and alters the transcriptome To investigate the effects of statin pleiotropy on endothelial function, we used an established protocol^[62]10 to generate iPSC-ECs from three healthy donors without familial hypercholesterolemia or CVDs ([63]Fig. 1a). These iPSC-ECs exhibited a typical ‘cobblestone’ morphology and expressed high levels of EC-specific markers, such as CD31 and vascular endothelial (VE)-cadherin ([64]Fig. 1b). We first tested the effects of seven statins, namely, pravastatin, atorvastatin, mevastatin, fluvastatin, lovastatin, rosuvastatin and simvastatin, at three concentrations (0.1 μM, 1 μM and 10 μM) on iPSC-EC functions. Because expressing high levels of NOS3 is a hallmark of functional ECs, we quantified its mRNA levels in vehicle- and simvastatin-treated iPSC-ECs and observed that simvastatin was the most potent statin that can upregulate NOS3 expression in a dose-dependent manner ([65]Fig. 1c and [66]Extended Data Fig. 1a). As doses of simvastatin beyond 10 μM compromised cell viability and increased reactive oxygen species (ROS) production in iPSC-ECs ([67]Extended Data Fig. 1b,[68]c), we chose 1 μM simvastatin for the following functional assays and transcriptomic and epigenetic profiling in iPSC-ECs. Consistent with the upregulated expression of NOS3, simvastatin improved the capacity of in vitro tube formation ([69]Fig. 1d and [70]Extended Data Fig. 1d) and nitric oxide (NO) production ([71]Fig. 1e) in iPSC-ECs compared to the vehicle control. Fig. 1 |. Simvastatin improves endothelial function by altering chromatin-associated transcriptome profiles of iPSC-ECs. Fig. 1 | [72]Open in a new tab a, Schematic diagram of the experimental design. iPSC-ECs differentiated from three healthy donors (D1, D2 and D3) were treated with statins for functional assays and multiomic analysis. CHIR, CHIR99021. b, Representative immunofluorescent images showing iPSC-ECs (iECs) from three healthy donors (D1, D2 and D3) expressing the typical endothelial surface markers CD31 (green) and VE-cadherin (red). Scale bar, 50 μm. c, Simvastatin exhibits a dose-dependent upregulation of NOS3 in iPSC-ECs (n = 4 biological samples). d, Simvastatin-treated iPSC-ECs showing a higher density of capillary-like tubes than the vehicle (DMSO)-treated counterparts (n = 3 biological samples). e, Simvastatin-treated iPSC-ECs produce a higher level of NO than the vehicle-treated counterparts (n = 3 biological samples). f, Transcriptomic analysis showing differential expression of genes in iPSC-ECs after treatment with simvastatin versus vehicle control. g,h, Enrichment analysis showing upregulated (g) and downregulated (h) DEGs in simvastatin-treated iPSC-ECs compared to the vehicle control (red, terms associated with vascular function; blue, terms associated with epigenetic regulation). rRNA, ribosomal RNA. i,j, Circle plots showing upregulated (i) and downregulated (j) DEGs under the indicated ontologies in simvastatin- versus vehicle-treated iPSC-ECs. All data are presented as mean ± s.e.m. Unpaired two-sided Student’s t-test (c–e). Next, we conducted next-generation sequencing analysis on vehicle- and simvastatin-treated iPSC-ECs. Compared to the vehicle, simvastatin upregulated 1,019 genes and downregulated 1,600 genes in iPSC-ECs ([73]Fig. 1f and [74]Extended Data Fig. 1e). The upregulated genes were enriched in processes such as angiogenesis, anti-inflammatory response and extracellular matrix (ECM) organization ([75]Fig. 1g). Notably, these gene ontology (GO) terms for biological processes are associated genes that enhance endothelial functions ([76]Fig. 1i): ETS1 and PPARA encode negative regulators of the inflammatory response^[77]11; NOS3, TIE1 and EPHB4 encode mediators of angiogenesis^[78]12; VWF and KDR are mature endothelial marker genes^[79]13; and KLF2 and KLF4 encode critical endothelial transcription factors (TFs) involved in endothelial biology and physiology^[80]14. By contrast, simvastatin downregulated genes such as HMGB2, CHAF1B, SET and EED, which are associated with epigenetic regulation of transcription, nucleosome assembly and chromatin organization ([81]Fig. 1h,[82]j). No overt differences in cell proliferation between vehicle- versus simvastatin-treated iPSC-ECs were observed ([83]Extended Data Fig. 1f). GO terms for cellular component revealed that most downregulated genes were associated with the nucleus and chromosomes ([84]Extended Data Fig. 1g,[85]h). Collectively, these results suggest that the beneficial effects of simvastatin on ECs might be achieved, in part, by modulating the activity of associated genes at the chromatin level. Simvastatin alters enhancer accessibility in iPSC-ECs While statins can regulate gene expression in cancer and atherosclerosis via epigenetic inhibition of histone deacetylase^[86]8,[87]15, how they alter the chromatin accessibility of target genes in vascular cells remains unknown. As we observed a significant downregulation of genes associated with epigenetic regulation in simvastatin-treated iPSC-ECs, we next conducted a genome-wide chromatin accessibility assessment in another set of vehicle- and simvastatin-treated iPSC-ECs using ATAC-seq. In line with our transcriptomic data ([88]Fig. 1h), we detected a reduced intensity of chromatin accessibility peaks (±5 kb of ATAC signal summit) in simvastatin- versus vehicle-treated iPSC-ECs ([89]Fig. 2a). Interestingly, our ATAC peak-annotation results showed that most of the accessible regions in iPSC-ECs were intergenic (36%) and intronic (45.7%), and 11% of ATAC peaks were promoters ([90]Fig. 2b). Therefore, most ATAC regions in iPSC-ECs were primarily associated with non-promoter regulation. To better understand the regulatory role of ATAC peak regions in iPSC-ECs, we next annotated our ATAC-seq data with the chromatin immunoprecipitation followed by sequencing (ChIP–seq) datasets of diverse histone marks in iPSC-ECs from the Encyclopedia of DNA Elements (ENCODE) ([91]Fig. 2c). Active enhancers are characterized by the concomitant presence of histone 3 lysine 4 (H3K4) monomethylation (H3K4me1) and histone 3 lysine 27 (H3K27) acetylation (H3K27ac), and poised enhancers harbor H3K27 trimethylation (H3K27me3) and H3K4me1 (ref. [92]16). Because our ATAC-seq peak regions in iPSC-ECs were substantially related to H3K27ac and H3K4me1 and devoid of H3K27me3 ([93]Fig. 2c), we infer that they are primarily located at active enhancer regions. Fig. 2 |. Simvastatin reduces chromatin accessibility at TEAD elements and represses YAP activity. Fig. 2 | [94]Open in a new tab a, Aggregated ATAC-seq signals across the peak summit (±5 kb) showing reduced accessibility of iPSC-ECs in the simvastatin group as compared to the vehicle control group (blue, vehicle control; red, simvastatin). CPM, counts per million. b, Annotation of all chromatin accessibility regions from the ATAC-seq data of simvastatin- and vehicle-treated iPSC-ECs. UTR, untranslated region. c, A heatmap showing the enrichment and depletion of various histone marks (from ENCODE; see [95]Methods for ENCODE accessions) at the peak regions of our ATAC-seq dataset. H3K4me3, H3K4 trimethylation; H3K36me3, histone 3 lysine 36 trimethylation. d, ATAC-seq footprinting heatmap showing global differential accessibility scores between control- and simvastatintreated iPSC-ECs (FDR < 0.1). e, Motif-enrichment analysis at the ATAC-seq sites identified KLFs and TEADs as the most predominantly affected motifs with differential chromatin accessibility. TEAD, TEAD1 (Homer unpublished data); TEAD1 (Homer encode data). f, ATAC-seq footprinting heatmap showing differential accessibility scores between control- and simvastatin-treated samples at loci marked with TEAD or KLF motifs (fold change (FC) > 0.5 and FDR < 0.01). g, KEGG pathway analysis of the ATAC-seq peaks with TEAD motifs. cAMP, cyclic AMP; RAP1, repressor activator protein; TRP, transient receptor potential. h, Representative immunofluorescent images showing reduced YAP nuclear localization in iPSC-ECs treated with simvastatin versus vehicle control. Blue, DAPI; green, YAP. Scale bar, 100 μm. i, Quantitative data showing nuclear/cytoplasmic YAP ratios in iPSC-ECs treated with simvastatin and vehicle control (n = 11 cells). j, Western blot analysis showing that simvastatin downregulated total YAP, SOX9 and CTGF protein levels in iPSC-ECs (n = 2 biological samples). k, Quantification of YAP, SOX9 and CTGF protein-level changes in simvastatin- and vehicle-treated iPSC-ECs. All data are presented as mean ± s.e.m. Unpaired two-sided Student’s t-test (i,k). Simvastatin reduces chromatin accessibility by blocking YAP activity Next, we investigated the chromatin accessibility of ATAC peaks globally and sought to identify the increased and decreased chromatin accessible regions caused by simvastatin treatment ([96]Fig. 2d). Unsupervised motif-enrichment analysis showed that the Krüppel-like factor and specificity protein (KLF and SP) family of TFs were the most significantly enriched motifs in regions with increased accessibility ([97]Fig. 2e,[98]f). By contrast, TFs in the TEAD family (that is, TEAD, TEAD1, TEAD3 and TEAD4) were the most significantly enriched motifs in regions with decreased accessibility ([99]Fig. 2e,[100]f). In line with our initial ATAC peak annotations ([101]Fig. 2b), when we annotated these ATAC peaks based on their increased or decreased accessibility, we observed that simvastatin-altered chromatin regions remained primarily enhancer regions rather than promoter regions ([102]Extended Data Fig. 2a). TEADs are broadly expressed TFs that regulate differentiation, development and tumorigenesis. However, little is known about their role in the vascular system^[103]17. As KLF and SP regulate vascular functions by binding to the promoters and enhancers of genes with similar DNA sequences^[104]14, we hypothesized that TEADs regulate endothelial functions through a similar mechanism. Indeed, after we binned ATAC signals into enhancer regions and performed motif analysis, we observed that the motifs that were associated with vascular endothelial growth factor (VEGF) signaling and endothelial function, such as ETV2, JUNB and ATF3, showed increased accessibility at enhancer regions ([105]Extended Data Fig. 2b)^[106]18. Importantly, consistent with our global ATAC-seq analysis ([107]Fig. 2e,[108]f), the TEAD motifs remained the most significantly enriched motifs with decreased accessibility at enhancer regions ([109]Extended Data Fig. 2c). Next, we performed Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of adjacent genes in the regions associated with TEAD and KLF motifs. Our data further confirmed that these motifs were enriched in pathways associated with mesenchymal cells such as Hippo and transforming growth factor (TGF)-β signaling ([110]Fig. 2g) and EC functions such as VEGF signaling ([111]Extended Data Fig. 2d), respectively. Similarly, GO pathway analysis revealed that simvastatin-altered accessibility regions were mainly associated with vascular functions, including angiogenesis and VEGF receptor signaling ([112]Extended Data Fig. 2d–[113]f). Collectively, our in-depth analysis of ATAC-seq data suggested that simvastatin exerts its pleiotropic effects on iPSC-ECs by reducing chromatin accessibility primarily at the enhancer regions associated with TEAD motifs. Although TEADs (TEAD1–TEAD4) can directly bind to DNA, they are not able to regulate gene expression independently due to the lack of transcription activation domains. When forming a complex in the nucleus, YAP and TEAD can activate target genes^[114]19. To determine the effect of simvastatin treatment on YAP activity in iPSC-ECs, we first assessed the subcellular distribution of YAP by performing immunofluorescence staining ([115]Fig. 2h,[116]i). At the baseline condition, both cytoplasmic and nuclear YAP were detectable in iPSC-ECs. However, simvastatin-treated iPSC-ECs showed a significant reduction in nuclear YAP, suggesting an inhibitory effect of simvastatin on YAP nuclear translocation. This finding was further supported by western blot results, which showed a significant decrease in total YAP protein levels ([117]Fig. 2j,[118]k). The major downstream proteins of YAP signaling, SOX9 and connective tissue growth factor (CTGF), were also downregulated by simvastatin in iPSC-ECs ([119]Fig. 2j,[120]k). In sum, our results suggest that simvastatin can decrease chromatin accessibility at TEAD elements through inhibiting YAP signaling in iPSC-ECs. YAP activity was previously shown to be negatively regulated by cell density^[121]20. Therefore, we performed comparative experiments by treating confluently and sparsely seeded iPSC-ECs with vehicle or simvastatin to assess their contributions to YAP activity. Simvastatin significantly downregulated YAP signaling in both confluent and sparse iPSC-ECs, as well as reduced YAP and SOX9 protein levels, independent of cell density ([122]Supplementary Fig. 1a–[123]d). Simvastatin also downregulated YAP in primary ECs ([124]Supplementary Fig. 2a–[125]d), supporting simvastatin’s inhibitory role in endothelial YAP signaling. Simvastatin inhibits TEAD–YAP activity To explore the kinetics of statin-induced transcriptome and epigenetic changes, iPSC-ECs treated with simvastatin for 0–72 h were subjected to both RNA-sequencing (RNA-seq) and ATAC-seq analyses. Principal component analysis (PCA) of RNA-seq data showed that simvastatin induced significant changes in global transcriptomic profiles ([126]Fig. 3a). Simvastatin-induced changes in global transcriptomic profiles were observed as early as 12 h and were sustained until the final examined time point at 72 h ([127]Extended Data Fig. 3a). KEGG pathway analysis showed that simvastatin upregulated the steroid biosynthesis pathway ([128]Fig. 3b). By contrast, simvastatin significantly downregulated the Hippo signaling pathway ([129]Fig. 3b) and concomitantly upregulated KLF2 and KLF4 and typical endothelial marker genes (NOS3, CDH5 and KDR) ([130]Fig. 3c and [131]Extended Data Fig. 3b). Genes downstream of the Hippo–YAP pathway^[132]21 were downregulated after simvastatin treatment ([133]Fig. 3d and [134]Extended Data Fig. 3c). Moreover, mesenchymal genes, such as TGFBR1, TWIST1, SNAI2 and SOX9, were also significantly downregulated by simvastatin ([135]Extended Data Fig. 3d). Fig. 3 |. RNA-seq and ATAC-seq analyses of simvastatin-treated iPSC-ECs at different time points. Fig. 3 | [136]Open in a new tab a, PCA plot of RNA-seq results of simvastatin-treated iPSC-ECs at 0 h, 12 h, 24 h and 72 h. PC, principal component. b, KEGG pathway analysis of RNA-seq data showing multiple dysregulated pathways, including the Hippo pathway, in iPSC-ECs after 72 h of treatment with simvastatin. c, Normalized expression levels of endothelial marker genes NOS3 and KLF2 from RNA-seq data of simvastatin-treated iPSC-ECs at different time points (n = 2 RNA-seq biological samples). d, Normalized expression levels of YAP downstream genes TGFB1 and FGF1 from RNA-seq data of simvastatin-treated iPSC-ECs at different time points. e, Heatmap of ATAC-seq data of simvastatin-treated iPSC-ECs at 0 h, 12 h, 24 h and 72 h. f, TEAD motifs were significantly downregulated in iPSC-ECs after simvastatin treatment for 12 h, 24 h and 72 h versus 0 h. Values indicate −log[10] (P values). NS, not significant. g, Schematic design of a YAP–TEAD activity fluorescence reporter system. CMV, cytomegalovirus; IRES, internal ribosome entry site. h, Representative fluorescent images of iPSC-ECs transfected with a YAP^GFP–TEAD^mCherry reporter before treatment with simvastatin and vehicle control. i, Schematic design of a YAP–TEAD activity luciferase reporter system. j, Simvastatin reduced TEAD activity in iPSC-ECs compared to vehicle control over the course of 72 h using a TEAD response element-driven luciferase reporter (n = 5 biological samples). All data are presented as mean ± s.e.m. One-way ANOVA corrected with the Bonferroni method (j). To assess the correlations between simvastatin-induced changes in chromatin accessibility and gene expression over the course of 72 h, we further analyzed ATAC-seq data generated from the same set of iPSC-ECs. Our data showed that the changes in chromatin accessibility were initiated after 12 h of simvastatin treatment, which became more striking at 24 h and 72 h ([137]Fig. 3e). Our time-resolved ATAC-seq data showed that 24 h of simvastatin treatment resulted in the most significant chromatin changes, such as downregulation of TEAD motifs ([138]Fig. 3e,[139]f). Moreover, we observed a strong concordance between chromatin accessibility and gene expression changes in iPSC-ECs after 24 h of simvastatin treatment ([140]Extended Data Fig. 4a). Collectively, these data strongly support a protective role of simvastatin in iPSC-ECs by modulating chromatin accessibility and gene expression patterns that confer improved EC functions. After observing a significant decrease in nuclear YAP immunofluorescence intensity in iPSC-ECs following 24 h of simvastatin treatment ([141]Extended Data Fig. 4b,[142]c), we further leveraged a reporter system comprising a YAP-driven green fluorescent protein (GFP) tag and a TEAD-responsive element-mediated nuclear histone 2B–mCherry tag to examine whether simvastatin can directly inhibit YAP–TEAD activity ([143]Fig. 3g)^[144]22. In the YAP–TEAD reporter iPSC-ECs, a robust nuclear TEAD signal at baseline ([145]Fig. 3h) was significantly diminished after 24 h of simvastatin treatment ([146]Extended Data Fig. 4d). We also found that simvastatin reduced TEAD activity after only 12 h of treatment using a TEAD response element-driven luciferase reporter ([147]Fig. 3i,[148]j). In sum, these results support a direct role for simvastatin in repressing YAP–TEAD activity by blocking nuclear YAP translocation in ECs. Simvastatin inhibits YAP by blocking geranylgeranyl pyrophosphate (GGPP)–Rho signaling Growing evidence suggests that the mevalonate pathway diverges at farnesyl pyrophosphate, which either generates squalene for cholesterol biosynthesis or gives rise to GGPP, which modulates Rho signaling^[149]23. Interestingly, statins were shown to reduce YAP activity in breast cancer cells by inhibiting the geranylgeranylation of RhoA^[150]24. Therefore, we hypothesized that simvastatin suppresses endothelial YAP activity by inhibiting GGPP–Rho signaling. Compared to the vehicle control, simvastatin upregulated KLF2 and downregulated VCAM1, a YAP target gene associated with inflammation, in iPSC-ECs ([151]Fig. 4a). However, mevalonic acid (MA), a product of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, reversed the expression patterns of these genes in the presence of simvastatin ([152]Fig. 4a). The addition of GGPP but not squalene in the presence of simvastatin resulted in a similar gene expression pattern for KLF2 and VCAM1 in iPSC-ECs, as did MA, suggesting that simvastatin improved endothelial functions by inhibiting GGPP. Simvastatin-induced reduction in the nuclear localization of YAP was reversed by MA and GGPP but not by squalene ([153]Fig. 4b,[154]c). While simvastatin enhanced tube-formation capacity, this effect was blunted by GGPP and MA but not by squalene ([155]Fig. 4d and [156]Extended Data Fig. 4e). MA and GGPP, but not squalene, abolished simvastatin-mediated improvements on endothelial NO release ([157]Fig. 4e). Fig. 4 |. Simvastatin improves endothelial functions through the GGTase–RhoA–YAP signaling axis. Fig. 4 | [158]Open in a new tab a, Quantitative PCR analysis showing expression levels of KLF2 and VCAM1 in iPSC-ECs treated with simvastatin, simvastatin + MA, simvastatin + GGPP, simvastatin + squalene, GGTi298, RhoAi and vehicle control (n = 3 biological samples). b, Representative immunofluorescent images showing YAP nuclear localization patterns (Cell Signaling Technology, 93622) in iPSC-ECs treated with simvastatin, simvastatin + MA, simvastatin + GGPP, simvastatin + squalene, GGTi298, RhoAi and vehicle control. Blue, DAPI; green, YAP. Scale bar, 100 μm. c, Nuclear/cytoplasmic YAP ratios in iPSC-ECs treated with simvastatin, simvastatin + MA, simvastatin + GGPP, simvastatin + squalene, GGTi298, RhoAi and vehicle control (n = 48 cells). d, Quantification of capillary-like tubular networks formed by iPSC-ECs treated with simvastatin, simvastatin + MA, simvastatin + GGPP, simvastatin + squalene, GGTi298, RhoAi and vehicle control (n = 3 biological samples). e, NO production by iPSC-ECs treated with simvastatin, simvastatin + MA, simvastatin + GGPP, simvastatin + squalene, GGTi298, RhoAi and vehicle control (n = 6 biological samples). f, Schematic overview of the regulation of YAP by the mevalonate pathway. Metabolites are shown in green boxes, and specific enzymes are also labeled. PP, pyrophosphate. All data are presented as mean ± s.e.m. One-way ANOVA corrected with the Bonferroni method (a,c–e). To further validate the causal role of simvastatin-mediated blockage of GGPP–RhoA signaling in YAP inhibition and improved endothelial functions, we used GGTi298, a specific GGTase inhibitor targeting the geranylgeranylation of RhoA, and a RhoA inhibitor (RhoAi) to treat iPSC-ECs. Indeed, both GGTi298 and RhoAi suppressed nuclear translocation of YAP ([159]Fig. 4b,[160]c), upregulated KLF2 ([161]Fig. 4a) and downregulated VCAM1 in iPSC-ECs ([162]Fig. 4a). Moreover, GGTi298 and RhoAi improved tube formation and NO release in iPSC-ECs ([163]Fig. 4d,[164]e and [165]Extended Data Fig. 4e). In sum, these results suggest that simvastatin improved iPSC-EC functions by inhibiting the GGTase–RhoA–YAP signaling axis ([166]Fig. 4f). Simvastatin represses YAP-dependent EndMT to improve EC function Endothelial dysfunction is an early manifestation of vascular dysfunction in T2D^[167]25. While T2D significantly increases the risk of developing coronary artery disease^[168]26, statins have been shown to effectively reduce major cardiovascular events in patients with T2D^[169]27. To understand the protective role of statins in endothelial dysfunction beyond their cholesterol-lowering effects, we cultured iPSC-ECs under diabetic conditions in vitro and sought to determine whether targeted inhibition of the GGTase–RhoA–YAP axis could rescue endothelial dysfunction. Specifically, iPSC-ECs from one male and one female donor were subjected to a diabetic milieu composed of high glucose, insulin and endothelin 1 (ET-1; ref. [170]28). While such an HG condition significantly impaired tube formation and NO production capacity in iPSC-ECs, these pathological phenotypes were improved by simvastatin, GGTi298 and RhoAi ([171]Fig. 5a–[172]c). Moreover, simvastatin, GGTi298 and RhoAi blunted the increase in ROS in iPSC-ECs maintained under the HG condition ([173]Extended Data Fig. 5a). Fig. 5 |. Simvastatin rescues diabetes-induced endothelial dysfunction. Fig. 5 | [174]Open in a new tab a, Representative images of capillary-like tube densities in iPSC-ECs under conditions of HG, HG + simvastatin, HG + GGTi298, HG + RhoAi and vehicle control. Scale bars, 250 μm. b, Quantification of capillary-like tube numbers in iPSC-ECs under conditions of HG, HG + simvastatin, HG + GGTi298, HG + RhoAi and vehicle control (n = 3 biological samples). D1, donor 1 iPSC-ECs; D2, donor 2 iPSC-ECs. c, NO production levels in iPSC-ECs under the control condition, the HG condition and that of HG + simvastatin reveal that simvastatin restores HG-impaired NO production (n = 3 biological samples). d, Transcriptome profiles of iPSC-ECs treated with HG, HG + simvastatin and vehicle control. e, Venn diagrams of transcriptomic analysis showing overlapping genes in HG versus vehicle control and HG versus HG + simvastatin. Top: overlapping upregulated genes. Bottom: overlapping downregulated genes. f, Gene set enrichment analysis plots of the EMT gene set in HG versus vehicle control (top) and HG versus HG + simvastatin (bottom). NES, normalized enrichment score, relative to vehicle control or HG + simvastatin. g, Heatmap of ATAC-seq data showing differential peaks in diabetic stress HG versus vehicle-treated iPSC-ECs. h, Motif analysis identifies both increased and decreased chromatin accessibility to the ATAC regions in HG-treated iPSC-ECs. i, KEGG pathway analysis shows dysregulated pathways in HG conditions compared to vehicle control-treated iPSC-ECs. HTLV-1, human T lymphotropic virus type 1; ARVC, arrhythmogenic right ventricular cardiomyopathy; AMPK, AMP-activated protein kinase. All data are presented as mean ± s.e.m. Unpaired two-sided Student’s t-test (b), one-way ANOVA corrected with the Bonferroni method (c). Next, we performed transcriptome analysis on iPSC-ECs exposed to the diabetic condition with or without simvastatin ([175]Fig. 5d). Remarkably, iPSC-ECs in both the vehicle and simvastatin–HG (HG + simvastatin) groups showed an almost 90% overlap of differentially expressed genes (DEGs) compared to those maintained under the HG condition, suggesting that simvastatin restored hyperglycemia-induced transcriptomic profiles toward the baseline condition ([176]Fig. 5e). We also observed a high degree of overlap in the top ten enriched gene sets between the vehicle and HG–simvastatin groups ([177]Extended Data Fig. 5b). Most of these gene sets were associated with metabolic pathways, such as glycolysis and mammalian target of rapamycin complex 1 signaling. Notably, an interesting gene set involved in the epithelial-to-mesenchymal transition (EMT) process was significantly activated in the HG group as compared to the vehicle and HG–simvastatin groups ([178]Fig. 5f). EMT, which refers to the EndMT process in ECs, contributes substantially to CVDs^[179]29. Intriguingly, we observed that simvastatin effectively repressed several clusters of genes with critical EndMT functions that were activated in iPSC-ECs under the diabetic condition ([180]Extended Data Fig. 5c). These clusters included ECM-producing genes such as COL1A1 and COL3A1; ECM modulator genes such as PCOLCE2, POSTN and FBN1; cell adhesion genes such as ITGB5, CDH2, VIM and THY1; EMT-associated signaling genes such as TGFB1, DKK1, WNT5A, PDGFRB and FGF2; and classic EMT regulator genes such as MEST, NID2, SNAI2 and PRRX1. In accordance with simvastatin-mediated downregulation of EndMT genes in iPSC-ECs cultured under the diabetic condition, our ATAC-seq data showed that chromatin accessibility of loci associated with critical endothelial TFs and TEADs changed significantly after 7 d of exposure to the diabetic condition ([181]Fig. 5g,[182]h). While simvastatin reduced YAP–TEAD activity, increased accessibility at chromatin associated with TEAD motifs suggested that YAP–TEAD was activated under the diabetic condition. Moreover, KEGG pathway analysis of the neighboring genes of the ATAC peaks revealed that the Hippo signaling pathway was significantly upregulated in HG-treated ECs ([183]Fig. 5i). We hypothesized that the rescue effects of simvastatin on T2D-induced endothelial dysfunction occur through the inhibition of YAP signaling. Indeed, we observed that simvastatin significantly reduced the protein levels of YAP and its downstream target SOX9, which were elevated under diabetic conditions ([184]Fig. 6a,[185]b and [186]Extended Data Fig. 5d,[187]e). Simvastatin, GGTi298 and RhoAi attenuated the accumulation of nuclear YAP induced by HG medium in iPSC-ECs ([188]Fig. 6c,[189]d). To further confirm the causal role of YAP inhibition in this process, we used CRISPR interference (CRISPRi) technology to knock down YAP expression ([190]Fig. 6e,[191]f). YAP repression via CRISPRi rescued diabetes-induced endothelial dysfunction in iPSC-ECs ([192]Fig. 6g). Moreover, the expression levels of VCAM1 and SOX9, two major YAP downstream genes, and SNAI2 and TWIST1, two classic EMT regulator genes, which were increased by HG medium, were restored to the baseline condition by simvastatin, GGTTi298, RhoAi and YAP repression ([193]Fig. 6h,[194]i). These data collectively support the idea that simvastatin protected endothelial function not only at the baseline level but also under diabetic conditions by suppressing the GGTase–RhoA–YAP axis. Fig. 6 |. Simvastatin rescues diabetes-induced endothelial dysfunction via inhibition of the YAP-dependent mevalonate pathway bifurcation. Fig. 6 | [195]Open in a new tab a, Western blot analysis showing that simvastatin downregulated total YAP and SOX9 levels in iPSC-ECs. b, Densitometric quantification of YAP and SOX9 protein expression changes in iPSC-ECs treated with vehicle control, HG medium, HG + simvastatin, HG + GGTi298 and HG + RhoAi (n = 2 biological samples). c, Representative immunofluorescent images showing YAP nuclear localization in iPSC-ECs treated with vehicle control, HG medium, HG + simvastatin, HG + GGTi298 and HG + RhoAi. Blue, DAPI; green, YAP. Scale bar, 100 μm. d, Nuclear/cytoplasmic YAP ratios in iPSC-ECs treated with vehicle control (n = 61 cells), HG medium (n = 58 cells), HG + simvastatin (n = 55 cells), HG + GGTi298 (n = 77 cells) and HG + RhoAi (n = 55 cells). e, Schematic design of CRISPRi to repress YAP1 transcription. Guided by a designed gRNA, CRISPRi uses a Tet-on controlled dCas9 (fused with a KRAB effector domain) to target the YAP1 promoter for transcriptional silencing upon addition of doxycycline. f, Quantitative PCR analysis showing significantly repressed YAP expression after targeted CRISPRi (n = 3 biological samples). g, Capillary-like tube numbers in iPSC-ECs transduced with the YAP CRISPRi (Tet-on dCas9) system in the vehicle control condition with dCas9 off, HG + dCas9 off and HG + dCas9 on (n = 3 biological samples). h, Quantitative PCR analysis showing the expression levels of VCAM1 and SOX9 in iPSC-ECs treated with diabetic medium (HG), HG + simvastatin, HG + GGTi298, HG + RhoAi and HG + YAP CRISPRi (n = 3 biological samples). i, Quantitative PCR analysis of SNAI2 and TWIST1 expression in iPSC-ECs treated with vehicle, HG and HG + YAP CRISPRi (n = 3 biological samples). All data are presented as mean ± s.e.m. Unpaired two-sided Student’s t-test (f), one-way ANOVA corrected with the Bonferroni method (b–d,g–i). Simvastatin rescues endothelial dysfunction in diabetic mice To investigate the protective effects of simvastatin and GGTi298 on endothelial function in vivo, we administered each compound separately to db/db mice, a mouse model of leptin-deficient T2D, via daily oral gavage for 8 weeks. We then assessed changes in vascular function using wire myography, as previously described^[196]10. After 8 weeks of treatment ([197]Fig. 7a), we dissected descending thoracic aortas from mice of all groups to evaluate changes in vascular tone ([198]Extended Data Fig. 6a). At the baseline level, the aortic rings isolated from db/db mice showed a lower basal endothelial-dependent relaxation response to acetylcholine than those from db/+ mice, indicating impaired endothelial function in the diabetic mice ([199]Fig. 7b). Simvastatin significantly improved vascular function by a complete restoration of acetylcholine-induced endothelial-dependent relaxation ([200]Fig. 7b, top), whereas GGTi298 exhibited a milder rescue effect ([201]Fig. 7b, bottom). Interestingly, we also observed a decreased basal contractile response to ET-1 in db/db mouse aortas, suggesting that endothelial-modified vascular smooth muscle cell function was also compromised ([202]Extended Data Fig. 6b). However, both simvastatin and GGTi298 effectively restored ET-1-induced vasoconstriction to the level observed in control mice ([203]Extended Data Fig. 6b). The protective role of simvastatin on vascular smooth muscle cell function might be attributed to its anti-inflammatory effects^[204]30. As neither simvastatin nor GGTi298 improved aortic vasoconstriction or vasodilation in wild-type (WT) control mice ([205]Extended Data Fig. 6c), our data collectively suggest that suppressing the GGTase–RhoA–YAP axis represents a promising strategy to potently rescue diabetes-induced endothelial dysfunction in vivo. Fig. 7 |. In vivo validation of the vasculoprotective role of simvastatin in a diabetic mouse model. Fig. 7 | [206]Open in a new tab a, Experimental design schematic showing four groups of mice with different treatments. b, Concentration–response relationship for acetylcholine (Ach)-induced endothelial-dependent aortic relaxation in mice treated with simvastatin (top) and GGTi298 (bottom). Each point represents the mean relaxation response ±s.e.m. (n = 3 biological samples). c, PCA showing that simvastatin- and GGTi298-treated db/db mice were close to non-diabetic control (db/+) mice. d, KEGG pathway analysis showing GGTi298- and simvastatin-repressed genes that were upregulated in db/db aortas. AKT, serine–threonine kinase; MAPK, mitogen-activated protein kinase; NF-κB, nuclear factor κB; PI3K, phosphoinositide 3-kinase; TNF, tumor necrosis factor. e, Schematic diagram of different regions of the aorta receiving different blood flow shear stress patterns. f, Representative brightfield images of iPSC-ECs under static, disturbed flow, disturbed flow + simvastatin, laminar flow and laminar flow + simvastatin conditions after 72 h. g, Nuclear/cytoplasmic YAP ratios in iPSC-ECs under conditions of disturbed flow (n = 117 cells), disturbed flow + simvastatin (n = 117 cells), laminar flow (n = 127 cells) and laminar flow + simvastatin (n = 127 cells). All data are presented as mean ± s.e.m. Two-way ANOVA corrected with the Bonferroni method (b). Unpaired two-sided Student’s t-test (g). *P < 0.05. Simvastatin represses YAP activity and EndMT in diabetic mice To better understand how simvastatin and GGTi298 alleviated diabetes-induced endothelial dysfunction in diabetic mice, we performed RNA-seq on aortic tissues collected from the same region for wire myography but from another set of mice ([207]Fig. 7a). PCA analysis showed that simvastatin- and GGTi298-treated db/db aortas were clustered together and closer to db/+ control aortas but separated from vehicle-treated db/db aortas ([208]Fig. 7c). Next, we analyzed the DEGs in db/db aortas treated with vehicle, simvastatin and GGTi298. Compared to those in vehicle-treated db/db aortas, 1,174 DEGs were found to be significantly restored by both simvastatin and GGTi298 ([209]Extended Data Fig. 7a). We divided the 1,174 DEGs into diabetes-upregulated and -downregulated categories to perform further KEGG pathway analyses. We found that diabetes-upregulated genes were significantly enriched in the Hippo signaling- and inflammation-associated pathways ([210]Fig. 7d), which were repressed by simvastatin and GGTi298 ([211]Extended Data Fig. 7b). By contrast, genes downregulated by diabetes linked to vascular functions, such as blood pressure, vasoconstriction and the oxidation–reduction process, were upregulated by simvastatin and GGTi298 ([212]Extended Data Fig. 7c). In line with our iPSC-EC data, simvastatin and GGTi298 rescued endothelial marker genes such as Klf2, Nr2f2 and Klf5 that were repressed in db/db aortas ([213]Extended Data Fig. 7d). Collectively, these data demonstrated that simvastatin and GGTi298 alleviated diabetes-induced endothelial dysfunction by repressing YAP-dependent pathways, such as EndMT. To examine whether differential shear flow patterns could affect YAP signaling along the aorta in the presence of simvastatin ([214]Fig. 7e), we applied disturbed (aortic arch, atheroprone) versus laminar flow (descending aorta, atheroprotected) to iPSC-ECs to mimic this physiological feature ([215]Fig. 7e). Specifically, iPSC-ECs treated with vehicle or simvastatin were concurrently exposed to laminar (12 dyne cm^−2) or oscillatory shear stress (±4 dyne cm^−2) for 72 h as previously reported^[216]31. Although iPSC-ECs aligned with the direction of laminar flow and showed chaotic alignment under disturbed flow ([217]Fig. 7f), simvastatin significantly reduced nuclear YAP signals under both flow patterns ([218]Extended Data Fig. 7e and [219]Fig. 7g). Moreover, simvastatin reduced a critical YAP downstream target, SOX9, in iPSC-ECs under both flow conditions ([220]Extended Data Fig. 8a,[221]b). Collectively, these data reaffirmed a causal role of simvastatin-repressed YAP signaling and EndMT in improved endothelial function. Simvastatin suppresses YAP–TEAD activity via a SOX9 enhancer We further investigated whether YAP can directly interact with EndMT genes in iPSC-ECs. We first conducted ChIP–seq analysis on simvastatin- and GGTi298-treated iPSC-ECs and examined YAP-binding signals at regions ranging from −5 kb to +5 kb from the peak summit. Both simvastatin and GGTi298 reduced YAP-binding signals ([222]Fig. 8a). Consistent with previous studies^[223]32, most of the YAP-binding regions (73.5%) in our ChIP–seq data were located at enhancer regions ([224]Fig. 8b). Simvastatin- and GGTi298-treated iPSC-ECs showed a more similar YAP-binding pattern than the control group ([225]Fig. 8c). TEAD motifs were the most enriched motifs in simvastatin- and GGTi298-treated iPSC-ECs ([226]Fig. 8d,[227]e). Interestingly, SOX9, a major cardiac fibrosis and EndMT TF^[228]33,[229]34, was identified in our motif analysis ([230]Fig. 8e). Next, we identified two major YAP-binding chromatin regions, chromosome (chr)17:71,249,620–71,250,020 and chr9:136,589,603–136,590,003, that were significantly downregulated upon simvastatin and GGTi298 treatment ([231]Fig. 8f,[232]g and [233]Extended Data Fig. 8c). Among the two chromatin regions, chr9:136,589,603–136,590,003 is located at the NOTCH1 enhancer region, whereas chr17:71,249,620–71,250,020 is part of the SOX9 enhancer region ([234]Fig. 8g). Notably, gene expression of both Sox9 and Notch1 was activated in db/db aortas compared to db/+ control aortas ([235]Extended Data Fig. 8d), suggesting that the two EndMT regulators may be the targets of statins in diabetic arteries. Fig. 8 |. Inhibition of the mevalonate pathway decreases YAP–TEAD binding at the SOX9 enhancer. Fig. 8 | [236]Open in a new tab a, Average YAP ChIP–seq enrichment scores among vehicle-, GGTi298- and simvastatin-treated iPSC-ECs (blue, vehicle control; orange, GGTi298; red, simvastatin). b, Annotation of ChIP–seq YAP-binding regions. TTS, transcription termination site. c, Unsupervised hierarchical clustering of vehicle-, GGTi298- and simvastatin-treated iPSC-ECs using the differentially enriched binding regions. d, ChIP–seq footprinting heatmap showing differential binding regions between iPSC-ECs treated with vehicle versus GGTi298 or simvastatin (FDR < 0.1). e, Overlapping YAP-binding motifs in iPSC-ECs treated with GGTi298 and simvastatin versus vehicle. f, Top decreased overlapping YAP-binding peaks in iPSC-ECs treated with GGTi298 and simvastatin versus vehicle. g, Genome Browser snapshots of the decreased binding loci (SOX9 enhancer) after simvastatin and GGTi298 treatment (highlighted in a yellow frame). KO, knockout. h, Immunostaining of endothelial markers, CD31 and VE-cadherin in SOX9 enhancer-knockout iPSC-ECs. i, Quantitative PCR analysis showing decreased levels of SOX9 in SOX9 enhancer-knockout cells (n = 3 biological samples). j, Quantitative PCR analysis of SOX9 gene expression in SOX9 enhancer-knockout and WT iPSC-ECs under diabetic conditions (HG) (n = 3 biological samples). k, Tube formation changes in HG-treated SOX9 enhancer-knockout and WT iPSC-ECs (n = 3 biological samples). All data are presented as mean ± s.e.m. Unpaired two-sided Student’s t-test (i–k). As Notch receptor 1 (NOTCH1) has binary roles in regulating endothelial functions by promoting angiogenesis and positively regulating the EndMT process during both physiological and pathological conditions^[237]35, we decided to focus on understanding the role of the previously undescribed SOX9 enhancer in simvastatin-mediated endothelial protective effects. We knocked out the SOX9 enhancer region using CRISPR–Cas9 in one iPSC line. The single-guide RNA (sgRNA) species were designed to target the upstream and downstream regions of chr17:71,249,620–71,250,020 ([238]Supplementary Fig. 3a), and a 540-bp fragment of the SOX9 enhancer was successfully excised ([239]Supplementary Fig. 3b,[240]c). We confirmed that knocking out the SOX9 enhancer in iPSCs did not compromise endothelial differentiation, as shown by high levels of EC marker genes ([241]Fig. 8h). Compared to parental control (WT) iPSC-ECs, SOX9 enhancer-knockout iPSC-ECs showed significantly downregulated SOX9 expression ([242]Fig. 8i) and did not respond to HG-induced upregulation of SOX9 nor to simvastatin-induced downregulation of SOX9 ([243]Fig. 8j). SOX9 enhancer-knockout iPSC-ECs also failed to respond to HG-induced upregulation and simvastatin-induced downregulation of an EndMT master gene, SNAI2 ([244]Supplementary Fig. 3d). By contrast, KLF2 expression was not compromised in SOX9 enhancer-knockout iPSC-ECs ([245]Supplementary Fig. 3e). Finally, simvastatin failed to restore diabetes-induced angiogenesis defects in SOX9 enhancer-knockout iPSC-ECs ([246]Fig. 8k). Overall, our data suggest that simvastatin improved endothelial function by epigenetically blocking YAP–TEAD-mediated upregulation of chromatin accessibility at EndMT-regulating genes by repressing the GGTase–RhoA–YAP–SOX9 signaling axis ([247]Extended Data Fig. 9). Discussion Endothelial dysfunction contributes to CVD-associated morbidity and mortality. In this study, we show in iPSC-ECs that YAP can translocate to the nucleus, where it binds TEAD to form a functional transcription complex that epigenetically upregulates genes associated with EndMT ([248]Extended Data Fig. 9). While sustained hyperglycemia can further activate YAP and EndMT to aggravate endothelial dysfunction, we showed that simvastatin effectively rescued this pathology by blocking the GGTase–RhoA–YAP axis. Cardiac-specific deletion of YAP decreased cardiomyocyte proliferation and led to myocardial hypoplasia, whereas endothelial-specific deletion of YAP compromised vascularization^[249]36,[250]37. However, YAP activation in adult tissues is generally believed to be pathogenic, as it was detected in mouse atherosclerotic plaques^[251]31, diabetic kidneys^[252]38 and diabetic cardiomyopathic hearts^[253]39. The YAP–TEAD complex has been reported to influence downstream gene transcription activity through regulating chromatin alterations at target loci^[254]40. Because ECs express higher levels of YAP than other cell types in the aorta^[255]41, endothelial YAP may maintain higher chromatin accessibility at target genes even under physiological conditions. Indeed, we observed that simvastatin improved endothelial functions not only under diabetic conditions but also at the baseline level. Because activated YAP can upregulate EndMT-regulating genes, it explains why ECs are prone to transitioning to a mesenchymal phenotype both in vitro and in vivo. Several studies showed that YAP activity was upregulated in HG conditions and T2D^[256]39,[257]42. We cross-validated the effectiveness of modulating simvastatin-mediated epigenetic repression of YAP signaling and EndMT in rescuing diabetes-induced endothelial dysfunction using simvastatin, GGTi298 and YAP CRISPRi. Additionally, we identified two previously undescribed enhancer regions (NOTCH1 and SOX9) as the direct binding sites of YAP. Using CRISPR technology, we further confirmed that the SOX9 enhancer is essential for the simvastatin-repressed EndMT process. Because our ChIP–seq data suggest that SOX9 motifs are also YAP targets, activated SOX9 may synergistically regulate downstream EndMT genes such as TWIST1, TWIST2, SNAI1 and SNAI2 with YAP^[258]34. EndMT has been recognized as therapeutically promising target for treating vascular dysfunction, and major EndMT drivers (for example, those encoded by SNAI1, SNAI2, TWIST1/2, ZEB1 and ZEB2) are controlled by epigenetic regulators^[259]43. Thus, our study provides an alternative strategy to modulate EndMT by epigenetically repressing the GGTase–RhoA–YAP–SOX9 signaling axis. Human iPSC-ECs offer tremendous potential for revealing pathogenic mechanisms and assessing drug responses in a patient-specific manner. However, several technical limitations, as outlined below, need to be addressed in future research to make this in vitro model a more effective platform for disease modeling and drug testing. First, iPSC-derived cells are less mature than their primary counterparts, which may lead to incomplete recapitulation of disease phenotypes observed in patients^[260]44. Second, the reprogramming process of iPSCs inevitably eliminates disease phenotype-associated epigenetic alterations, making it challenging to investigate patient-specific disease phenotypes and drug responses caused by complex interactions of multiple genetic mutations and environmental risk factors^[261]45. Nevertheless, with the advance of bioengineering tools in promoting cell maturation and the emergence of multiomic studies focusing on identification of diabetes-associated variants in large-scale populations^[262]46, iPSC-ECs derived from diabetic patients will be a useful platform to decode the effects of patient-specific epigenetic changes on endothelial dysfunction. In summary, our findings uncover a previously unidentified interaction between statin-mediated chromatin remodeling and improved endothelial function via repressing the GGTase–RhoA–YAP–SOX9 signaling axis. The causal role of YAP activation in EndMT and vascular dysfunction could lead to the development of alternative biomarkers for CVD diagnosis and therapeutic strategies to prevent or treat endothelial dysfunction-associated CVDs. Methods Human iPSC reprogramming and culture Human iPSCs were generated from peripheral blood mononuclear cells collected from healthy donors using the CytoTune-iPS 2.0 Sendai reprogramming kit (Thermo Fisher Scientific). In brief, peripheral blood mononuclear cells were isolated from 20 ml of whole blood using Ficoll-Paque Premium (GE HealthCare) and cultured for 9 d in blood medium. A total of 1 × 10^6 cells were transfected with the Sendai reprogramming virus overnight. Cells were then transferred to a mouse embryonic fibroblast-coated plate and cultured in mTeSR medium (Stemcell Technologies). Human embryonic stem cell-like colonies were picked after day 20, and iPSC clones were transferred to Matrigel-coated plates (BD Biosciences) for expansion. Human iPSCs were passaged using Accutase (Global Cell Solutions) and 10 μM Y-27632 (Selleckchem) and then refreshed with daily changes of mTeSR medium. Two clones of iPSCs from each donor were maintained and passaged at the same time. The maintenance medium for iPSCs was switched to chemically defined E8 medium (Gibco) after passage 3. The E8 medium was changed daily, and iPSCs were passaged when they reached >80% confluency. Aliquoted iPSCs were cryopreserved between passages 15 and 30 for downstream experiments. For human iPSC reprogramming, written consents were obtained from these individuals, and the studies were approved by the Stanford Administrative Panels on Human Subjects Research. EC differentiation Endothelial differentiation was carried out in RPMI medium with B27 supplement and no insulin (Gibco) when iPSCs (at passages 15–30) reached 80–90% confluency. In brief, iPSCs were treated with 6 μM CHIR99021 (Selleckchem) for 2 d and then 2 μM CHIR99021 for another 2 d. Starting from day 4, the differentiation medium was changed to Endothelial Cell Growth Medium-2 (EGM-2) medium (Lonza) supplemented with VEGF (50 ng ml^−1, PeproTech), basic fibroblast growth factor (20 ng ml^−1, PeproTech) and the TGF-β inhibitor SB431542 (10 μM, Selleckchem). On day 12, differentiated iPSC-ECs were enriched using CD144 microbeads (Miltenyi Biotec). Sorted iPSC-ECs were cultured in gelatin-coated plates using EGM-2 medium. Confluent iPSC-ECs were passaged using TrypLE (Gibco) and cryopreserved at passage 2 after derivation using Bambanker (Wako Chemicals). To account for technical variations (for example, culture medium, coating matrix and growth factors) on transcriptome profile changes, two clones of each iPSC line were differentiated into ECs in two independent batches. Transcriptomic analysis and functional experiments were conducted using recovered iPSC-ECs. Drug and compound treatment iPSC-ECs were seeded at 30,000 cells per well in six-well plates or 1,500 cells per well in 96-well plates depending on which experiment was carried out. The following drugs and compounds were used in this study: simvastatin (0.1–10 μM; Sigma-Aldrich, S6196), lovastatin (0.1–10 μM; Sigma-Aldrich, PHR1285), atorvastatin (0.1–10 μM; Sigma-Aldrich, PZ0001), rosuvastatin (0.1–10 μM; Sigma-Aldrich, SML1264), mevastatin (0.1–10 μM; Sigma-Aldrich, M2537), fluvastatin (0.1–10 μM; Sigma-Aldrich, SML0038), pravastatin (0.1–10 μM; Sigma-Aldrich, P4498), GGTi298 (1 μM; Sigma-Aldrich, G5169), (±)-MA 5-phosphate (0.5 mM, 1 μM; Sigma-Aldrich, 79849), GGPP (20 μM; Sigma-Aldrich, G6025), squalene (20 μM; Sigma-Aldrich, S3626) and RhoAi C3 (2 μg ml^−1; Cytoskeleton, CT-04). Unless otherwise specified in the figure legends, iPSC-ECs were treated with drugs for 72 h. Quantitative real-time PCR (rtPCR) Total RNA from iPSC-ECs was isolated using the RNeasy Plus Mini Kit (Qiagen), and reverse transcription was performed using the High-Capacity cDNA Reverse Transcription Kit (Life Technologies). Quantitative rtPCR reactions were carried out using TaqMan Universal PCR Master Mix (Applied Biosystems) according to the manufacturer’s protocol in a CFX96 real-time PCR detection system (Bio-Rad). Relative mRNA levels were normalized against glyceraldehyde 3-phosphate dehydrogenase (GAPDH) in each reaction. TaqMan primers and probes were ordered from Thermo Fisher Scientific (NOS3, Hs01574659_m1; KLF2, Hs00360439_g1; VCAM1, Hs01003372_m1; SNAI2, Hs00161904_m1; TWIST1, Hs01675818_s1; and SOX9, Hs00165814_m1). More than three biological replicates per group were performed. In vitro model of hyperglycemia iPSC-ECs were seeded at 30,000 cells per well in six-well plates or 1,500 cells per well in 96-well plates according to different experimental settings. To mimic the diabetic condition in vitro, cells were maintained in EGM-2 medium supplemented with 25 mM d-glucose (equivalent to 4.5 mg ml^−1 blood glucose in patients), 20 nM insulin and 10 nM ET-1 for 7 d with daily medium changes. Simvastatin, GGTi298 and RhoAi were introduced to the diabetic condition for 72 h to rescue endothelial dysfunction. Mannitol (25 mM) was used in the vehicle control group to normalize osmolality. Immunofluorescence staining and quantification iPSC-ECs were seeded onto gelatin-coated eight-chamber glass slides (Nunc) and maintained in EGM-2 medium. Cells were fixed using 4% PFA after 2 d of culture and then permeabilized with 0.1% Triton X-100 (Sigma-Aldrich) and blocked with 2.5% donkey serum (Sigma-Aldrich) in PBS. Cells were then incubated with primary antibodies (anti-CD31 (PECAM), R&D Systems; anti-VE-cadherin, R&D Systems; anti-YAP, Novus Biologicals; YAP, Cell Signaling Technology) in 2.5% donkey serum–PBS overnight at 4 °C. After three washes with PBS, cells were further incubated with secondary antibodies (donkey anti-mouse Alexa Fluor 488; donkey anti-rabbit Alexa Fluor 594, Life Technologies) at room temperature for 1 h. Next, slides were washed with PBS and mounted with SlowFade Gold Antifade Reagent with 4,6-diamidino-2-phenylindole (DAPI; Life Technologies) before imaging. All immunofluorescent images were captured using a Nikon Eclipse 80i fluorescence microscope and analyzed using ImageJ. The intensity of YAP-positive signals in the nuclear versus cytoplasmic fractions was analyzed using CellProfiler software. Confluent and sparse iPSC-EC culture Five times more iPSC-ECs were seeded in the confluent condition (5,000 cells per well) compared to the sparse condition (1,000 cells per well) in 96-well plates, and simvastatin was administered to both conditions at the same time. Cells were subjected to immunofluorescence staining and western blot analysis for YAP signaling. Cell viability assay Differentiated iPSC-ECs were seeded on gelatin-coated 96-well plates at 10,000 cells per well. Cells were treated with simvastatin at 0 μM, 0.1 μM, 0.3 μM, 1 μM, 3 μM and 10 μM for 48 h. Cell viability after simvastatin treatment was evaluated using a CellTiter-Glo viability assay kit (Promega) according to the manufacturer’s procedures. Briefly, a volume of CellTiter-Glo reagent equal to the volume of culture medium was added to each well. The cells and reagents were mixed for 2 min on an orbital shaker. Next, the cells were lysed for another 10 min to stabilize the luminescent signal. Raw readings were recorded using a Cytation 5 plate reader or imager (BioTek Instruments). Cell proliferation measurement The cell-proliferation assay was performed by tracking cell number changes over the course of simvastatin treatment for 3 d. In brief, cells were seeded in the same condition, and replicates were fixed every day during the 3-d treatment with simvastatin and dimethyl sulfoxide (DMSO). All fixed cells were stained with DAPI and counted to assess simvastatin’s effect on cell proliferation. ROS measurement iPSC-ECs in different treatment groups were maintained in 96-well white-walled plates with clear bottoms (Falcon). ROS-Glo H[2]O[2] substrate solution (Promega) was prepared according to the manufacturer’s instructions. H[2]O[2] substrate solution (20 μl) was added to each well 2 h before the end of each treatment. The final volume in each well (H[2]O[2] substrate solution plus culture medium) was 100 μl, and plates were incubated at 37 °C in an incubator with 5% CO[2]. Next, 50 μl of supernatant was combined with 50 μl of ROS-Glo H[2]O[2] Detection solution in a separate opaque white plate. After 20 min of incubation at room temperature, the relative luminescence units were measured using the GloMax-Multi detection plate reader (Promega). In vitro tube formation assay iPSC-ECs were seeded at 40,000 cells per well in a Matrigel (growth factor reduced; Corning)-coated 24-well plate. Cells in each treatment group were further cultured in EGM-2 medium with 50 ng ml^−1 VEGF overnight at 37 °C. Images were taken, and tubular knots were quantified to compare the in vitro angiogenesis capacity of iPSC-ECs with different treatments. NO measurement NO production in iPSC-ECs was measured with the Nitrate/Nitrite Fluorometric Assay Kit (Cayman Chemical). In brief, iPSC-ECs from different donors were seeded at the same density in gelatin-coated six-well plates and cultured for 3 d until the cells reached over 80% confluency. Next, cells were treated with 10 μM acetylcholine chloride (Sigma) overnight, and the supernatant was collected and stored at −80 °C. The thawed supernatant was centrifuged to remove cell debris before adding nitrate reductase to convert nitrate to nitrite. Total nitrite was measured using a Cytation 5 Cell Imaging Multimode Reader (BioTek). The total NO concentration for each sample was calculated using a standard curve measured at the same time with samples. YAP CRISPRi construct, virus production and transfection The sgRNA sequence targeting the YAP promoter was ligated into the pSLQ1373 vector with BstXI and Xhol (YAP CRISPRi sgRNAs: GCCGCCGCCAGGGAAAAGAA, GGCCTTCGCAGCCCCCGCAG). Lentivirus was produced using pSLQ1373 containing YAP sgRNA and packaging protein vectors (pMD2.G and pCMV-dR8.91). In brief, HEK293T cells were transfected with these three plasmids using TransIT-LT1 transfection reagents (Mirus). Lentiviral supernatants were collected after 48 h of transfection and used for transduction of an iPSC line containing a Tet-On 3G system driving dead Cas9 (dCas9)–Krüppel-associated box (KRAB) expression^[263]47. For YAP CRISPRi experiments, the puromycin-purified YAP CRISPRi iPSC line was differentiated into iPSC-ECs, and doxycycline was added into the iPSC-EC culture medium to turn on dCas9–KRAB expression. Transcription-repression efficiency was measured by quantifying the mRNA level of YAP using quantitative PCR. Generation of a YAP–TEAD reporter line The YAP–TEAD reporter plasmid (pLL3.7 FLAG-YAP1-TEAD-P-H2B-mCherry) was purchased from Addgene (plasmid 128327). The lentiviral YAP–TEAD reporter was packaged into HEK293T cells using packaging vectors (pLL3.7 FLAG-YAP1-TEAD-P-H2B-mCherry, pMD2.G and pCMV-dR8.91), which were transduced into iPSCs (D1 iPSC line) using 5 μg ml^−1 polybrene (Sigma-Aldrich) overnight. The iPSCs carrying the YAP–TEAD reporter were purified using GFP selection and expanded for iPSC-EC differentiation for experiments. Similarly, a TEAD response element-driven luciferase reporter (BPS Bioscience) was transduced into iPSC-ECs as a lentivirus using 5 μg ml^−1 polybrene overnight. Next, an equal number of transfected iPSC-ECs was seeded into 96-well plates (5,000 cells per well), followed by treatment with vehicle control (DMSO) or simvastatin. Cells were collected for TEAD luciferase-activity analysis at 12 h, 24 h and 72 h after treatment using a ONE-Step Luciferase Assay (BPS Bioscience). Laminar flow and disturbed flow experiments To mimic the effects of disturbed (aortic arch, atheroprone) versus laminar (descending aorta, atheroprotected) flow at different aortic regions on YAP signaling in the presence of simvastatin, iPSC-ECs were cultured under ibidi fluidic units (ibidi) according to the manufacturer’s instructions. In brief, 1 × 10^5 iPSC-ECs were plated on each μ-Slide I 0.4 Luer slide, which was attached in series to each fluidic unit with pumping EGM-2 medium. iPSC-ECs treated with vehicle or simvastatin were concurrently exposed to laminar shear stress (12 dyne cm^−2) and oscillatory shear stress (disturbed, ±4 dyne cm^−2) for 72 h. Slides were then collected for immunostaining and western blot analysis at the end of the experiment. CRISPR knockout experiments SOX9 enhancer-knockout sgRNAs were designed using a Synthego design tool and synthesized by Synthego. For electroporation, 3.5 × 10^5 iPSCs were transfected with 1 μl Cas9 protein (Integrated DNA Technologies, 20 μM) and 1 μl sgRNA (100 μM) in Resuspension Buffer R (Thermo Fisher) in a final volume of 10 μl. Cells were transfected with the Neon Transfection System (Thermo Fisher) following the electroporation procedure of 200 V, 20 ms and two pulses and then resuspended in E8 medium supplemented with 10 μM Y-27632 for 24 h. The SOX9 enhancer primers and OneTaq DNA Polymerase (NEB) were used to amplify the targeted loci following the manufacturer’s instructions. After PCR amplification, electrophoresis on a 2% agarose gel was used to characterize the PCR products. The QIAquick PCR Purification Kit (Qiagen) was used to purify the PCR products, which were then examined using Sanger sequencing (sgRNA: top, AUUUUCCCACAGAGGUAAUG; bottom, AUUGCCGCAAAGGAGCUUUG; Genomic PCR primers: ACGTCTCTTGCTTCCCAAACA, TGGTTTGAGTTTGTTTTCTTCTTCA). Subcellular fractionation and western blot analysis Cells were lysed on ice using RIPA buffer (Thermo Fisher) containing a protease inhibitor and phosphatase inhibitor cocktail (Sigma). Supernatants were collected by centrifugation at 14,000g for 20 min at 4 °C. The Subcellular Protein Fractionation Kit for Cultured Cells (Thermo Scientific) was used to separate cytoplasmic and nuclear proteins. Protein concentrations were quantified using a Pierce^™ BCA protein assay kit (Thermo Scientific). Equal quantities of protein lysates (25 μg) were loaded on NuPAGE Bis-Tris Gels (Life Technologies), and resolved bands were transferred to PVDF membranes using the Trans-Blot Turbo Transfer System (Bio-Rad). PVDF membranes were incubated with primary antibodies (anti-YAP, 1:1,000, Novus Biologicals; anti-phospho-YAP, 1:1,000, Cell Signaling Technology; anti-lamin A/C, 1:500, Santa Cruz; anti-SOX9, 1:1,000, Cell Signaling; anti-CTGF, 1:1,000, Thermo Scientific; anti-histone H3, 1:1,000, Cell Signaling Technology) overnight at 4 °C, followed by incubation with HRP-conjugated secondary antibodies (1:5,000) for 1 h at room temperature and then detected using the ChemiDoc XRS+ System (Bio-Rad). An HRP-conjugated GAPDH antibody (Life Technologies) was used as a loading control for normalization. Animals Six-week-old male C57BL/KsJ-db/db (db/db), non-diabetic (db/+) and WT mice (Jackson Laboratory) were used for experiments. No statistical methods were used to pre-determine sample size. Because men have a higher prevalence of T2D and benefit more from statin therapy in the management of cardiovascular risks than do women, to reduce variability, we used male mice and similar sample sizes based on our previous vascular animal studies^[264]48. This study was approved by the Stanford Administrative Panel on Laboratory Animal Care under protocol APLAC 33803, and the mice were housed in temperature-controlled environments. The drugs were dissolved in saline and administered through oral gavage once a day for 8 weeks. Specifically, five non-diabetic control (db/+) mice received saline as a control group, five db/db mice received saline as a vehicle control, five db/db mice received simvastatin treatment (25 mg per kg), and five db/db mice received GGTi298 treatment (5 mg per kg). Descending thoracic aortas from all groups were collected for a wire myography study and RNA-seq analysis at the end of treatment. We further included WT (C57BL/6) mice to test the effect of vehicle, simvastatin and GGTi298 on vascular function using wire myography. Isometric tension studies on mouse descending thoracic aortas After 8 weeks of statin and GGTi298 treatment, the aortas from mice of all groups were collected, cleaned of excess connective tissue and fat, cut into ~2-mm rings and mounted on two parallel tissue supports in the vessel incubation chamber filled with KHB buffer oxygenated continuously with 95% O[2]–5% CO[2] at 37 °C (KHB buffer composition in mM: 131.5 NaCl, 5 KCl, 1.2 NaH[2]PO[4], 2.5 MgCl[2], 2.5 CaCl[2]; 2 g glucose and 2 g NaHCO[3] in 1 L with pH 7.4). Aortas were subjected to isometric tension studies, and the developed force traces were recorded by LabChart software (ADInstruments). After two pre-constriction treatments with the vasoconstrictor PGF2-α (1 μM), the aortic rings were equilibrated for 30 min under a resting optimal tension of 10 mN with the chamber buffer changed every 10 min. The optimal resting tension was verified in each set of experiments before generating concentration–response curves. ET-1 (0.1 nM to 1 μM) was added to induce a contractile response. At the plateau of vasocontraction, acetylcholine (1 nM to 1 μM) was added accumulatively to induce vasorelaxation. Relaxation responses were calculated as the percent reduction in tension from the pre-contracted state. Contractile responses were calculated as the force generated by aortic rings calibrated by the dry weight with units of mN mg^−1. Control responses were measured by adding the same amount of vehicle (water). Transcriptomic analysis of AmpliSeq and RNA-seq data RNA was extracted from frozen iPSCs, iPSC-ECs and mouse aortas using the Qiagen RNeasy kit according to the manufacturer’s instructions. For AmpliSeq, 10 ng of RNA was used for cDNA synthesis with the SuperScript VILO cDNA Synthesis Kit (Thermo Fisher Scientific). The barcoded libraries were prepared with the Ion AmpliSeq Transcriptome Human Gene Expression Kit (Thermo Fisher Scientific) according to the manufacturer’s instructions. Combined libraries (50 pM) were loaded on Ion PI v3 chips using the Ion Chef automated platform (Thermo Fisher Scientific). AmpliSeq sequencing data were analyzed using the Ion Torrent Mapping Alignment Program (TMAP) as described in a previous study. To achieve both specificity and sensitivity, TMAP implemented a two-stage mapping approach with four alignment algorithms, BWA-short and long, SSAHA and Super-maximal Exact Matching, which were followed by the Smith–Waterman algorithm to find the final best mapping. RNA-seq analysis was conducted through Novogene. DEG analysis was performed with the R package DESeq2, which is available from Bioconductor. Genes with false discovery rate (FDR) or adjusted P value <0.05 are identified as DEGs. To identify potential function-enriched pathways, DEGs were analyzed for biological functions and pathway enrichment using the KEGG Orthology-Based Annotation System and gene set enrichment analysis. ATAC-seq and ChIP–seq data collection and analysis For ATAC-seq samples, 50,000 iPSC-ECs treated with simvastatin or vehicle control were collected. The transposition reaction was conducted using Nextera Tn5 Transposase. Transposed DNA was purified and amplified according to Buenrostro’s protocol^[265]49. For ChIP–seq samples, vehicle-, simvastatin- and GGIT298-treated iPSC-ECs were collected to perform chromatin immunoprecipitation using an anti-YAP antibody (Novus Biologicals) with the Zymo-Spin ChIP Kit (Zymo Research). Sheared chromatin was cross-linked (a small portion was saved as the ChIP–seq input sample) and incubated with anti-YAP antibody-conjugated Dynabeads overnight in a cold room for immunoprecipitation. Immunoprecipitated DNA was eluted, de-cross-linked and purified following the instructions of the Zymo-Spin ChIP Kit to prepare ChIP–seq libraries. ATAC-seq sequencing reads in FASTQ format were controlled for quality and trimmed with trim_galore in paired-end mode. The trimmed reads were aligned to the human genome (hg38) using Burrows–Wheeler Aligner version 0.7.17 (BWA-MEM). Duplicate reads were subsequently removed with Picard MarkDuplicates. Peak calling for each ATAC-seq replicate was performed using macs2 (version 2.2.7.1) callpeak with the options ‘–nomodel –shift −37 –extsize 73 –SPMR’. The quality of ATAC-seq peaks was assessed with the R package ATACseqQC. For ChIP–seq, peak calling was performed using DFilter with the parameters ‘-ks=10 -bs=50 -lpval=6 -wig -nonzero -refine’. The signal was converted into bigWig format, and visual inspection of peak quality was performed using the WashU Epigenome Browser. To identify differential accessible peaks, the R package DiffBind was employed using the de-duplicated alignment file in BAM format and peaks called by macs2 as inputs. The consensus peak was determined by including peaks that were called in more than two of the replicates. Affinity binding matrices were computed with DiffBind. Differential accessibility analyses were carried out using DESeq2. Genomic annotation of peaks and differential motif analyses were performed with Homer2. Genomic cis regulatory functional prediction analyses of differential peaks were performed using GREAT with basal plus extension association rules. Statistical analysis Statistical analyses were performed using Student’s t-test or one-way analysis of variance (ANOVA) corrected with the Bonferroni method. Multiple-group comparisons were conducted using two-way ANOVA corrected with the Bonferroni method. Sequencing data analysis was conducted with the Wald test and corrected for multiple testing using the Benjamini and Hochberg method. All error bars are defined as s.e.m. unless otherwise indicated. Statistical analysis was conducted on data from three or more biologically independent experimental replicates; western blot experiments were performed with two biological replicates. Extended Data Extended Data Fig. 1 |. Statins improve endothelial function and alter epigenetic associated genes in iPSC-ECs. Extended Data Fig. 1 | [266]Open in a new tab a, NOS3 expression levels in iPSC-ECs after being treated with seven statins at different concentrations (0.1 μM, 1 μM, and 10 μM). Data are normalized to that of the vehicle control group; prava, pravastatin; atorva, atorvastatin; rosuva, rosuvastatin; meva, mevastatin; fluva, fluvastatin; lova, lovastatin; and simva, simvastatin (n = 6 biological samples). b, Simvastatin decreases the viability of iPSC-ECs at 10 μM (n = 3 biological samples). c, Simvastatin increases ROS production in iPSC-ECs at 10 μM (n = 3 biological samples). d, Representative images showing higher densities of capillary-like networks formed by iPSC-ECs from three healthy donors treated with simvastatin versus vehicle. Scale bars, 250 μm. e, Differentially expressed genes (DEGs) in simvastatin- versus vehicle-treated iPSC-ECs (FDR < 0.05). f, Simvastatin had no effect on iPSC-EC proliferation compared to vehicle (DMSO) (n = 4 biological samples). g, Enrichment (cellular components) analysis of upregulated DEGs shown in e. h, Enrichment (cellular components) analysis of downregulated DEGs shown in (e). All data are presented as mean ± SEM. Unpaired two-sided Student’s t-test (a,b,c). Extended Data Fig. 2 |. Simvastatin alters chromatin accessibility in iPSC-ECs. Extended Data Fig. 2 | [267]Open in a new tab a, Annotation of ATAC peaks with differential chromatin accessibility. b and c, Motif enrichment analysis at the ATAC-seq sites with differential chromatin accessibility at enhancer regions. d, KEGG pathway analysis of the ATAC-seq peaks with KLF motifs. e and f, GO pathway analysis of ATAC-seq peaks with KLF motifs and TEAD motifs. Extended Data Fig. 3 |. RNA-seq and ATAC-seq analyses of iPSC-ECs treated with simvastatin at different timepoints. Extended Data Fig. 3 | [268]Open in a new tab a, Heatmap of RNA-seq showing changes in transcriptomic patterns of iPSC-ECs after being treated with simvastatin at 0 h, 12 h, 24 h, and 72 h. b, Normalized expression levels of endothelial marker genes (KLF4, CDH5, and KDR) from RNA-seq of simvastatin-treated iPSC-ECs at different timepoints. c, Normalized expression levels of YAP downstream genes (SMAD3, CTGF, GLI2, MCL1, RUNX1, BIRC2, TGFB2, and BIRC5) from RNA-seq of simvastatin-treated iPSC-ECs at different timepoints. d, Normalized expression levels of mesenchymal genes (TGFBR1, TWIST1, SNAI2, and SOX9) from RNA-seq of simvastatin-treated iPSC-ECs at different timepoints. All data are presented as mean ± SEM. n = 2 RNA-seq biological samples. Extended Data Fig. 4 |. Simvastatin inhibits YAP and TEAD activity in iPSC-ECs. Extended Data Fig. 4 | [269]Open in a new tab a, Comparative analysis showing that simvastatin exposure to iPSC-ECs for 12 h, 24 h, and 72 h induces highly correlated changes in DEGs and ATAC signal alterations (values indicate log[2](fold change)). b, Immunofluorescence of YAP subcellular localization in iPSC-ECs treated with simvastatin versus vehicle control for 24 h. c, Quantification of nuclear/cytoplasmic YAP ratios in iPSC-ECs treated with simvastatin versus vehicle control for 24 h (n = 14 cells). d, iPSC-ECs treated with simvastatin (n = 101 cells) but not vehicle (n = 138 cells) for 24 h showing significantly decreased nuclear TEAD activity. e, Representative images of capillary-like tubular networks formed by iPSC-ECs treated with simva, simva + MA, simva + GGPP, simva + squalene, GGTi298, RhoAi, and vehicle control. All data are presented as mean ± SEM. Unpaired two-sided Student’s t-test (c,d). Extended Data Fig. 5 |. Simvastatin reverses hyperglycemia (HG)-induced endothelial dysfunction by repressing YAP-mediated EndMT process. Extended Data Fig. 5 | [270]Open in a new tab a, ROS levels in iPSC-ECs treated with HG, HG + simvastatin, HG + GGTi298, HG + RhoAi, and vehicle control (n = 3 biological samples). b, Gene set enrichment analysis (GSEA) of the differentially regulated genes in HG versus vehicle control (top panel) and HG versus HG + simvastatin (bottom panel) reveals that HG-upregulated EndMT process is blunted by simvastatin. c, Heatmaps of epithelial-mesenchymal-transition gene sets in HG versus vehicle control (left) and HG versus HG + simvastatin (right) based on GSEA analysis. Representative western blot data (d) and densitometric quantification (e) showing the hyperglycemic condition (HG) decreases phosphorylated/total YAP in iPSC-ECs compared to the control condition. GAPDH serves as a loading control (n = 3 biological samples). All data are presented as mean ± SEM. Unpaired two-sided Student’s t-test (a). Extended Data Fig. 6 |. In vivo validation of the endothelial protective effects of simvastatin in a diabetic mouse model. Extended Data Fig. 6 | [271]Open in a new tab a, A representative trace of isometric tension in the mouse aorta. The aortic rings were equilibrated for 30 min under a resting tension of 10 mN after two sessions of pre-constriction with the vasoconstrictor PGF2α (1 μM). For a vasoconstriction response, endothelin-1 (ET-1, 0.1 nM to 1 μM) was used to induce a contractile response. At the plateau of maximal contraction, acetylcholine (Ach, 1 nM to 1 mM) was added accumulatively to initiate relaxation. b, Concentration-response relationship for ET-1-induced aortic constriction in mice treated with simvastatin (left panel) and GGTi298 (right panel). Developed tension was the force generated by aortic rings normalized to aortic tissue dry weight (mN/mg). Each point represents the mean developed contractile force ± SEM (n = 4 biological samples). c, Concentration-response relationship for ET-1-induced aortic constriction in wildtype mice treated with vehicle (saline), simvastatin (25 mg/kg), or GGTi298 (5 mg/kg) for 8 weeks (left panel). Concentration-response relationship for Ach-induced endothelial-dependent aortic relaxation in wildtype mice treated with vehicle (saline), simvastatin (25 mg/kg), or GGTi298 (5 mg/kg) for 8 weeks (right panel). Each point represents the mean constriction/relaxation response ± SEM (n = 4 biological samples). All data are presented as mean ± SEM. Extended Data Fig. 7 |. RNA-seq analysis shows potent rescue effects of both simvastatin and GGTi298 on diabetes-induced vascular dysfunction in mice. Extended Data Fig. 7 | [272]Open in a new tab a, Venn diagram showing overlapped DEGs of db/+ (vehicle), db/db + simvastatin, and db/db + GGTi298 compared to the db/db + vehicle group, respectively. b, EndMT-associated genes, such as Ctgf, Vcam1, Tgfb2, and Smad3, were rescued by simvastatin and GGTi298 in diabetic mouse aortas (n = 2 RNA-seq biological samples). c, KEGG pathway analysis showing both simvastatin and GGTi298 improved genes associated with endothelial functions that were downregulated in db/db mouse aortas. d, Endothelial marker genes, such as Klf2, Nr2f2, and Klf5, were restored by simvastatin (simva) and GGTi298 in db/db mouse aortas (n = 2 RNA-seq biological samples). e, Representative immunofluorescent images showing YAP nuclear localization patterns in iPSC-ECs treated with disturbed flow, disturbed flow + simvastatin, laminar flow, and laminar flow + simvastatin. Blue: DAPI. Green: YAP. Scale bars: 50 μm. All data are presented as mean ± SEM. Extended Data Fig. 8 |. Simvastatin-regulated enhancers identified by YAP ChIP-seq. Extended Data Fig. 8 | [273]Open in a new tab a, Western blot analysis showing simvastatin downregulated SOX9 expression in iPSC-ECs under disturbed and laminar flow patterns. b, Densitometric quantification of SOX9 protein expression changes in simvastatin- and vehicle-treated iPSC-ECs under disturbed and laminar flow patterns (n = 2 biological samples). c, Genome Browser snapshots of decreased binding loci at the NOTCH1 enhancer region showing simvastatin and GGTi298 treatment inhibited YAP binding at this region (highlighted in a yellow frame). d, Normalized gene expression levels (from RNA-seq) of Notch1 and Sox9 in aortas (n = 2 RNA-seq biological samples) from diabetic (db/db) and control mice (db/+). All data are presented as mean ± SEM. Unpaired two-sided Student’s t-test (b,d). Extended Data Fig. 9 |. Schematic summary demonstrates that simvastatin rescues endothelial dysfunction by repressing YAP-mediated chromatin remodeling of the EndMT process. Extended Data Fig. 9 | [274]Open in a new tab A schematic overview showing our proposed model. In normal endothelial cells (ECs), geranylgeranyltransferase (GGTase) mediates YAP activity through the mevalonate pathway. Active YAP creates an ‘open chromatin status’ at the enhancer regions of genes regulating EndMT, such as SOX9. The diabetic condition further enhances YAP activity, thereby exacerbating endothelial functions by further upregulating EndMT genes. In contrast, statins decrease mevalonate levels via the inhibition of HMG-CoA reductase, followed by suppression of GGTase-mediated RhoA geranylgeranylation, consequently attenuating YAP activity (dashed line). Reduced YAP activity makes chromatin less-open (‘closed status’) at the enhancer regions of genes associated with EndMT. Diabetes-induced endothelial dysfunction can be alleviated by suppressing YAP activity with statins, GGTi298 or RhoA inhibitor (RhoAi), leading to the downregulation of EndMT genes. P, phosphorylation. Supplementary Material Supplementary Information [275]NIHMS1923558-supplement-Supplementary_Information.pdf^ (8.1MB, pdf) Acknowledgements