Graphical abstract graphic file with name fx1.jpg [53]Open in a new tab Highlights * • RSAD2 as a pathogenic interferon-stimulated gene at the maternal-fetal interface * • RSAD2 excess increases lipid load in structural cell populations * • RSAD2-induced lipid accumulation impairs placental vasculogenesis * • LCA targeting of RSAD2 alleviates lipid-induced vascular inflammation __________________________________________________________________ The role of type I interferon response at the maternal-fetal interface is complex. Ding et al. identify RSAD2 as a pathogenic ISG in SLE pregnancies, where it drives placental lipid accumulation and impairs vasculogenesis, with LCA targeting RSAD2 to improve pregnancy outcomes. Introduction The prolonged and excessive induction of type I interferon (IFN-I) signaling impairs embryonic development,[54]^1^,[55]^2^,[56]^3 which is observed in some autoimmune diseases such as systemic lupus erythematosus (SLE) and Aicardi–Goutières syndrome.[57]^4^,[58]^5^,[59]^6 Upon binding to their receptor, IFN-I, mainly the multiple subtypes of IFN-α and IFN-β, induces hundreds of interferon-stimulated genes (ISGs), which function to restrict pathogenic infections and regulate the IFN response.[60]^7^,[61]^8^,[62]^9 However, the functions of ISGs in different diseases could be complex. Therefore, identifying the protective and pathogenic ISGs in these processes is essential for the accurate diagnosis and treatment of aforementioned diseases. Several protective ISGs are required for a successful pregnancy, whereby it plays a role in pregnancy recognition and uterine artery remodeling during pregnancy.[63]^10^,[64]^11 Previous studies have shown impaired decidual spiral artery remodeling in mice lacking the IFN-I receptor (Ifnar^−/− mice), indicating that activation of the IFN-I signaling pathway is crucial for early decidualization.[65]^12 Unlike the deciduomata of pseudopregnant mice, the presence of an embryo in the decidua of pregnant mice upregulated the expression of many ISGs, including Irf8, Iarp, Isg15, and Ifi27.[66]^13 Recently, a spatial multiomics map of trophoblast development in human early pregnancy showed that the transformation of interstitial extravillous trophoblast cells into placental bed giant cells, the receptors of the IFN-I signaling pathway, and the corresponding target, IFI27, are upregulated.[67]^14 This also suggests that a series of ISGs play a protective role in promoting embryo implantation, trophoblast invasion, and vascular remodeling. By contrast, excessive IFN-I signaling increases the activation of IFN-induced transmembrane proteins (IFITMs), which leads to the inhibition of placental syncytiotrophoblast formation and ultimately, fetal demise.[68]^2 Moreover, the inactivation of G protein-coupled estrogen receptor 1 contributes to the upregulation of canonical ISGs such as Ifit2, Isg15, and Ccl5, halting fetal development and promoting fetal demise during maternal inflammation.[69]^15 Considering that some ISGs exert protective effects during pregnancy, the identification of pathogenic ISGs could improve the treatment accuracy of IFN-I-related complications during pregnancy. SLE is an autoimmune disease characterized by the excessive production of IFN-I, especially IFN-α.[70]^16^,[71]^17^,[72]^18^,[73]^19 An ISG expression analysis of peripheral blood mononuclear cells (PBMCs) isolated from patients with SLE revealed that the expression of many key ISGs, including ISG15, ISG20, MX1, MX2, IFIT1, IFIT2, IFIT3, IFITM1, IFITM2, and RSAD2 (also called cytomegalovirus-inducible gene 5 [Cig5] and Cig33), was significantly activated.[74]^20^,[75]^21^,[76]^22 It is ∼7–9 times more prevalent in women than in men, especially in women of childbearing age.[77]^23^,[78]^24 Furthermore, elevated IFN-α activity early in gestation is associated with the onset of preeclampsia in patients with SLE, which occurs via the inhibition of angiogenesis.[79]^25^,[80]^26 In summary, the excessive activation of IFN-α signaling pathway is a high-risk factor for adverse pregnancy outcomes in SLE. Although specific guidelines for the management of SLE in pregnancy have been implemented, with hydroxychloroquine and aspirin treatment being recommended, recent data show that pregnancy outcomes in SLE have not improved over the past three decades.[81]^27^,[82]^28 Nevertheless, studies of the mechanisms by which IFN-α and its downstream pathogenic ISGs mediate adverse pregnancy outcomes in patients with SLE and those with other IFN-α-related pregnancy diseases are still very limited. Here, we demonstrated that RSAD2 is a pathogenic ISG at the maternal-fetal interface in patients with SLE, which causes lipid accumulation in the placenta, inhibited angiogenesis, and impaired fetal development. The small molecular compounds targeting RSAD2 alleviate abnormal pregnancy outcomes (APOs) in the SLE mouse model. Our work will contribute to the development of new treatment strategies for pregnant patients with a dysregulated IFN-I response. Results RSAD2 is highly expressed at the maternal-fetal interface in patients with SLE To identify potentially pathogenic ISGs in SLE pregnancy, we monitored ISGs expression in the PBMCs, decidua, and chorionic villi from pregnant patients with SLE using the quantitative real-time PCR array platform. We found that the expression of RSAD2 in PBMCs of pregnant patients with SLE was higher than in those from the group of healthy pregnant individuals ([83]Figures 1A and [84]S1A). The results indicated that the IFN-I response in the peripheral blood of pregnant patients with SLE was more activated than that of the control group. Next, we detected the expression of the indicated ISGs in situ in the decidua and chorionic villi and found that the expression of RSAD2 was also higher in these tissues than in those of the normal pregnant group ([85]Figure 1B). To investigate the response of the maternal-fetal microenvironment to IFN-α, we tested the induction of ISG expression in decidual and trophoblast organoids by treating them with IFN-α or phosphate-buffered saline (PBS, as a negative control). We found that the expression of RSAD2 was strongly induced by IFN-α in trophoblast organoids and decidual organoids ([86]Figures 1C and [87]S1B). We found that IFNAR1 was expressed in situ in the decidua and chorionic villi, as well as in the decidual and placental trophoblast-like organoids ([88]Figures 1D and 1E). To explore how various cell types responded to IFN-α, we sorted decidua-derived immune cells, including CD56^+ natural killer (NK) cells, CD3^+ T cells, and CD14^+ macrophages, as well as structural cells such as decidual stromal cells (DSCs) and trophoblasts. Gating strategy for immune cells was showed in [89]Figure S1C. We then detected ISG expression changes in these cells in response to IFN-α. We found that the structural cell populations at the maternal-fetal interface (i.e., DSCs and trophoblasts) mounted a strong IFN-α response ([90]Figures 1F and [91]S1D). Among the immune cells, macrophages were the most responsive to IFN-α, followed by T cells and NK cells ([92]Figure 1F). The high expression of RSAD2 in DSCs and trophoblast cells has also been confirmed at the protein level ([93]Figures 1G and 1H). The aforementioned results indicate that the structural cell population of the maternal-fetal interface is more capable of upregulating the expression of RSAD2 in response to IFN-I. Figure 1. [94]Figure 1 [95]Open in a new tab RSAD2 is highly expressed in pregnant patients with SLE (A) Heatmap showing the expression of interferon-stimulated genes (ISGs) in the peripheral blood mononuclear cells (PBMCs) of healthy women (Healthy PBMCs), female patients with SLE (SLE PBMCs), healthy pregnant women (Healthy pregnancy PBMCs), and pregnant patients with SLE (SLE pregnancy PBMCs). (B) Heatmap showing the expression of ISGs in the decidua and villi of healthy early pregnant women and pregnant patients with SLE. (C) Heatmap showing the expression of ISGs in the decidual and trophoblast organoids from the first trimester decidual gland or trophoblast cells (passage 1, day 5), treated with 1,000 U/mL IFN-α or PBS for 12 h. (A, B, and C). The 2^−ΔCt values of ISGs were calculated from the ISG qPCR array data. (D) Representative IHC image of IFNAR1 and CD55 (expressed in DSCs) co-staining of decidual tissue sections (top), and representative IHC image of IFNAR1 and EPCAM (expressed in VCTs, the proliferative villous cytotrophoblast cells) co-staining of chorionic villi sections (bottom); scale bar, 50 μm. (E) Representative IHC image of decidual organoids (top) from the first trimester decidual glands (passage 1, day 5) and of trophoblast organoids (bottom) from the first trimester trophoblast cells (passage 1, day 5); scale bar, 50 μm. (F) Expression of ISGs in decidual CD56^+ NK cells, CD3^+ T cells, CD14^+ macrophages, decidual stromal cells (DSCs), and trophoblasts (n = 3); the 2ˆ^−ΔΔCt values of ISGs were calculated from the qPCR data. (G and H) Western blot showing RSAD2 and ISG15 expression in DSCs and trophoblasts, treated with 1,000 U/mL IFN-α or PBS for 12 h. Data were analyzed using the unpaired t test and are presented as the mean ± SEM. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001; ns, not significant. RSAD2 upregulation is accompanied by accumulation of lipid droplets and elevated levels of proinflammatory prostaglandin To explore the role of RSAD2 in a high IFN-α environment at the maternal-fetal interface, we performed immunoprecipitation (IP) experiments targeting RSAD2 protein in DSCs and identified the IP products by liquid chromatography-tandem mass spectrometry (LC-MS/MS) ([96]Figure 2A). RSAD2 and some binding partners of it are typically associated with the mitochondria and lipid droplets (LDs) ([97]Figure 2B), which suggested that it could potentially alter the metabolism of the maternal-fetal interface. To test the ability of RSAD2 to regulate lipid metabolism in DSCs and trophoblasts, we analyzed the relationship between the RSAD2 expression and LD accumulation. Lipid metabolism was assessed in 293T and JEG3 (human placental choriocarcinoma) cells following the overexpression of RSAD2 through transduction with an RSAD2-overexpressing lentiviral vector or a negative control vector. RSAD2 overexpression efficiency was quantified by qPCR ([98]Figures 2D and 2F). LD staining showed that high RSAD2 expression in both 293T and JEG3 cells was associated with increased LD accumulation ([99]Figures 2C and 2E). In addition, we found that IFN-α treatment induced high RSAD2 expression in DSCs, which was accompanied by the marked accumulation of LDs; by contrast, with PBS treatment, DSCs lacking RSAD2 expression had only basal levels of LDs ([100]Figures S2A and S2B). We confirmed that RSAD2 expression and the accumulation of LDs were also observed in trophoblastic organoid models treated with IFN-α ([101]Figures S2D and S2E). To further characterize the effect of RSAD2 on the formation of LDs in the decidua and chorionic villi, we performed immunohistochemistry (IHC) on frozen sections from PBS- or IFN-α-treated tissue samples, and LDs were detected using oil red O staining. We observed an increase of LD level in the decidua ([102]Figure S2C), although it was only weakly increased in the chorionic villi under the same conditions ([103]Figure S2F). The aforementioned results show that RSAD2 upregulation is associated with increased LD concentration in DSCs and trophoblasts. To further test whether the RSAD2 overexpression is associated with lipid metabolism, we measured the expression levels of the major transcriptional regulators sterol regulatory element-binding protein and the endoplasmic reticulum-resident diacylglycerol acyltransferase 1; the genes encoding the fatty acid synthase, FASN, which catalyzes de novo lipid synthesis, and CD36, which assists in fatty acid uptake from the exogenous environment; and the genes encoding lipid transporter proteins, including apolipoprotein (apo) A1, APOA4, APOB, and APOD. The expression levels of the genes encoding these proteins were increased in RSAD2-overexpressing cell types ([104]Figures 2D and 2F) and IFN-α-treated trophoblasts than in those treated with PBS ([105]Figure S2G). Moreover, cells stably expressing RSAD2 displayed an elevated ability to take up fatty acids ([106]Figures S2H and S2I). To further confirm the downstream genes of RSAD2, the expression levels of RSAD2 were knocked down in 293T cells that had been overexpressed using small interfering RNA (siRNA) interference assays, which resulted in the downregulation of lipid metabolism genes ([107]Figure S2L). The effective siRNAs targeting RSAD2 were screened by qPCR and western blot ([108]Figures S2J and S2K). LD development has marked effects on the production of inflammatory mediators, including prostaglandin E2 (PGE2), interleukin (IL)-1β, and IL-6, in foamy macrophages.[109]^29 We found that the level of proinflammatory PGE2, PGI2, and PGD2 was significantly increased in DSCs in response to IFN-α ([110]Figures 2G and 2H). Taken together, our data indicate that RSAD2 is associated with lipid metabolic reprogramming at the maternal-fetal interface accompanied by LD accumulation and elevated levels of production of proinflammatory mediators. Figure 2. [111]Figure 2 [112]Open in a new tab Upregulation of RSAD2 is associated with inflammatory lipid accumulation at the maternal-fetal interface (A) Decidual stromal cells (DSCs) were treated with 1,000 U/mL IFN-α (+) or PBS (−) for 12 h. Cell lysates were subjected to immunoprecipitation (IP) with a rabbit anti-RSAD2 monoclonal antibody (mAb) or a rabbit IgG control. RSAD2 IP was determined by western blotting (upper). Heatmap of proteins interacting with RSAD2, identified using LC-MS/MS (lower). (B) Sankey map of cell localizations and functions of RSAD2-interacting proteins. (C) Representative IF image of LDs in 293T cells, which has been overexpressed RSAD2; scale bar, 10 μm. (D) qPCR analysis of lipid metabolism-related genes in RSAD2-overexpressing 293T cells. (E) Representative IF image of LDs in JEG3 cells, which has been overexpressed in RSAD2; scale bar, 10 μm. (F) qPCR analysis of lipid metabolism-related genes in RSAD2-overexpressing JEG3 cells. Data are representative of three independent experiments with similar results (C, D, E, and F). (G) Schematic diagram of the prostaglandin synthesis pathway. (H) DSCs from 12 donors were isolated and either treated with PBS or IFN-α for 12 h. To obtain enough cells for LC-MS/MS assay, the cells from every two donors were mixed as one sample. The threshold for identifying significant differences was VIP ≥ 1 and t test p < 0.05 in the orthogonal partial least squares discriminant analysis (OPLS-DA) model. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001; ns, not significant. To confirm that the high expression of RSAD2 is associated with the accumulation of LDs at the maternal-fetal interface in vivo, we employed a mouse pregnancy model in which high IFN-I levels were induced by poly(I:C) treatment at E10.5 ([113]Figure S3A). We found that poly(I:C) treatment induced a severe reduction in dam weight ([114]Figure S3B) and APOs ([115]Figures S3C–S3G). Intrauterine growth restriction in pups of the poly(I:C)-treated group was observed at E11.5 ([116]Figure S3C). The body weights, lengths, and widths of the live fetuses and the diameters of their placentas were also significantly smaller compared with the controls ([117]Figures S3D–S3G). We found that the murine decidua and placenta expressed significantly higher levels of RSAD2 and ISG15 following poly(I:C) treatment compared with those of control mice ([118]Figures 3A–3C and [119]S3H). Moreover, BODIPY 493/503 staining showed that LDs accumulated in the decidua and placenta after the intraperitoneal injection of poly(I:C), which was not observed in the control animals ([120]Figures 3D, 3E, and 3F). Oil red O staining confirmed that an increase in RSAD2 levels associated with excessive lipid accumulation ([121]Figures 3G, 3H, and 3I). Meanwhile, LC-MS/MS revealed that the triacylglycerol (TAG)56:6(16:0) and diacylglycerol (DAG) (16:0/18:1) content of the decidua and placenta, respectively, were increased in the poly(I:C)-treated mice compared with the controls ([122]Figures 3J and 3K). We performed an RNA sequencing (RNA-seq) analysis of saline- or poly(I:C)-treated decidua and placenta collected at E12.5 and found that the expression of the apolipoprotein (Apo) D-encoding gene, Apod, was significantly increase in the decidua ([123]Figure S3I), while that of Apoa1, Apoa4, and Apob was increased in the placenta ([124]Figure S3J). The significantly increased expression of a series of Apo genes at the maternal-fetal interface suggested that lipid metabolism was markedly perturbed. Collectively, our results showed that the upregulation of Rsad2 is associated with lipid accumulation at the maternal-fetal interface in the placenta. Figure 3. [125]Figure 3 [126]Open in a new tab Elevated RSAD2 expression is accompanied by lipid accumulation in the placenta (A) Rsad2 and Isg15 mRNA from the decidual and placental samples were analyzed with quantitative real-time PCR (n = 3). (B) Western blot analysis of RSAD2 and ISG15 protein expression in decidual and placental samples. The experiment has been independently repeated at least five times. (C) Data statistics of RSAD2 relative to tubulin expression were obtained by Image Lab. (D and E) LD staining of the decidua (D) and placenta (E) of pups from three independent pregnant dams at gd12.5. Images are representative of three independent experiments with similar results. Scale bar, 50 μm. (F) Quantification of LD staining by ZEN 2.6. (G and H) Oil red O staining of the decidua (G) and placenta (H) of pups from independent pregnant dams at gd12.5. Scale bar, 50 μm. Images are representative of three independent experiments. (I) Quantification of oil red O staining by Image-Pro Plus 6.0. (J) Heatmap representing the relative abundances of lipid subspecies. Each column represents a single decidual or placental sample from independent pregnant dams, and each row corresponds to a single subspecies of lipid. (K) Quantification of TAG56:6 (16:0) and DAG (16:0/18:1) subspecies. Poly(I:C)-treated decidual and placental samples are shown as red bars, and saline-treated samples are depicted as blue bars. All the data are presented as mean values ± SEM. p values were computed using the multiple t tests. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001; ns, not significant. RSAD2 depletion alleviates DAG-lipid accumulation in the placenta and adverse pregnancy outcomes To demonstrate that RSAD2 is a pathogenic ISG at the maternal-fetal interface, we next used Rsad2^−/−mice, which were generated using CRISPR-Cas9 technology, to determine the effect of RSAD2 on pregnancy outcomes ([127]Figure S4A). There were no obvious histopathological changes in the liver, spleen, and kidneys of Rsad2^−/−mice, compared with wild-type (WT) animals ([128]Figure S4B). We found that poly(I:C) treatment significantly reversed the phenotype of embryo absorption (the loss of embryos or fetuses, typically characterized by the absence of visible viable embryos or the presence of only residual blood clots in place of what would normally be a complete embryo) in Rsad2^−/−pregnant mice compared with WT pregnant mice at E12.5 ([129]Figures 4A and 4B). Specifically, compared with poly(I:C)-treated WT mice, embryonic developmental abnormalities and the embryo absorption rate significantly decreased in Rsad2^−/−pregnant mice ([130]Figures 4B and 4C). Moreover, the body lengths and the body widths of live fetuses were significantly smaller in poly(I:C)-treated WT mice compared with saline-treated controls. By contrast, the fetuses of the poly(I:C)-treated Rsad2^−/−mice were similar in size to those of the saline-treated control group ([131]Figure 4D). Moreover, we observed the following abnormalities in the poly(I:C)-treated WT mice but not in the Rsad2^−/−group: decidual vessel wall thickening, a reduction in the size of placental vascular spaces, and a decrease in the fetal blood cell content in the labyrinthine zone ([132]Figure S4C). The aforementioned results suggest that Rsad2, as a pathogenic ISG, induces adverse pregnancy outcomes. Figure 4. [133]Figure 4 [134]Open in a new tab RSAD2 depletion alleviates poor pregnancy outcomes and DAG-lipid accumulation (A) Representative image of pups from poly(I:C)- or saline-treated pregnant WT or Rsad2^−/−mice at gd12.5. Scale bar, 1 cm. (B) One representative litter from each of the indicated treatment groups collected at gd12.5 is shown. Scale bar, 1 mm. (C) Pie charts indicating the total number of fetuses analyzed and their developmental phenotypes (i.e., resorbed or halted development versus phenotypically normal), related to (A). WT-saline (n = 5), Rsad2^−/−-saline (n = 5), WT-poly(I:C) (n = 11), and Rsad2^−/−-poly(I:C) (n = 8) were enrolled. (D) The lengths (left) and widths (right) of live fetuses were measured with vernier calipers. WT-saline (n = 4), Rsad2^−/−-saline (n = 4), WT-poly(I:C) (n = 5), and Rsad2^−/−-poly(I:C) (n = 4) were measured. p values were computed using unpaired t test. (A), (B), (C), and (D) are representative of at least two independent experiments. (E and G) (E) LD staining and (G) oil red O staining of the deciduas and placentas from poly(I:C)-treated pregnant WT or Rsad2^−/−mice and saline-treated pregnant WT mice, collected at gd12.5. Images are representative of three independent experiments. Scale bar, 50 μm. DB, decidual basalis; LZ, labyrinth zone. (F) Quantification of mean fluorescence intensity (MFI) of LD staining in the labyrinth zone by ZEN 2.6. (H) Quantification of oil red O staining in the labyrinth zone by Image-Pro Plus 6.0. (F and H) Data are presented as the mean ± SEM and were obtained from the placentas of independent pregnant dams. p values were computed using the one-way analysis of variance (ANOVA). (I) Heatmap showing the relative abundances of lipid subspecies. Each column represents a single decidual or placental sample from independent WT or Rsad2^−/−pregnant dams (poly[I:C]-treated), and each row corresponds to a single subspecies of lipid. (J) Gene set enrichment analysis (GSEA) of genes associated with the triacylglyceride synthesis pathways expressed in the decidual samples of Rsad2^−/−mice compared to those expressed in the decidual samples of WT mice (poly[I:C]-treated). (A–J) Rsad2^−/−and WT mice were injected intraperitoneally with poly(I:C) or saline at gd10.5, and the tissues were harvested at gd12.5. All the data are presented as mean values ± SEM. (D) p values were computed using the multiple t tests. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001; ns, not significant. To explore whether high Rsad2 expression caused fetal demise in mice by promoting LD accumulation in the placenta, whole placenta frozen sections were stained with BODIPY 493/503. We found that, following poly(I:C) treatment, unlike that mass LD accumulation was observed in the WT animals, the LD content of the decidua and labyrinthine zone in Rsad2^−/−mice was similar with saline-treated WT mice ([135]Figures 4E and 4F). In addition, oil red O staining showed that large LDs were present in the decidua and placenta of WT mice following poly(I:C) treatment, while the lipid content of Rsad2^−/−mice was similar to that of saline-treated WT controls ([136]Figures 4G and 4H). The widely targeted quantitative lipidomics results revealed that the levels of DAG (16:0/18:1) and other sn-1,2-DAG species were significantly lower in the Rsad2^−/−placenta than those in WT placenta under the same poly(I:C) treatment ([137]Figure 4I). Although no significant changes in the decidual lipidome were observed in the poly(I:C)-treated Rsad2^−/−mice and WT mice ([138]Figure 4I), transcriptome data revealed that the TAG synthesis pathway was inhibited in Rsad2^−/− animals compared with poly(I:C)-treated WT mice ([139]Figure 4J). It has been reported that the sn-1,2-DAG-mediated activation of protein kinase Cε is implicated in obesity-related lipid metabolism disorders, where it induces hepatic insulin resistance and type Ⅱ diabetes.[140]^30 Moreover, the excessive accumulation of lipids leads to macrophage activation and the production of proinflammatory PGE2. In accordance, we found COX-2^+ macrophages in the placenta of poly(I:C)-treated WT mice but not in those of poly(I:C)-treated Rsad2^−/−mice or saline-treated WT animals ([141]Figure S4D). Taken together, our results show that Rsad2 is a pathogenic ISG, which causes adverse pregnancy outcomes by inducing DAG-lipid accumulation in the placenta. RSAD2 depletion alleviates placental vascular injury caused by DAG-lipid accumulation The fetus-derived placenta is composed of a layer of trophoblast giant cells, spongiotrophoblast layer, and a labyrinthine zone, the site of metabolic exchange between the maternal and fetal blood. The labyrinthine zone contains nonnucleated maternal red blood cells (RBCs) and fetal blood spaces, which are populated by nucleated fetal RBCs ([142]Figure 5A). Our earlier findings led us to hypothesize that the high expression of RSAD2 attenuated blood vessel development in the placenta by inducing excessive lipid accumulation. To test this hypothesis, we stained frozen placenta sections of saline- or poly(I:C)-treated WT mice with cytokeratin to label trophoblasts and BODIPY 493/503 probes to label LDs. We found that LDs mainly accumulated in the placental labyrinth ([143]Figure 5B) and partially colocalized with trophoblasts ([144]Figure 5C). This result suggests that lipid accumulation in the placenta damaged cells other than trophoblasts. Figure 5. [145]Figure 5 [146]Open in a new tab RSAD2 depletion alleviates placental vascular injury caused by lipid accumulation (A) Schematic diagram showing the decidual and placental architecture and the localization of different cell types within these structures. (B and C) OCT-embedded frozen placental sections from poly(I:C)- or saline-treated pregnant WT mice were stained for BODIPY 493/503 (used to stain LDs, green). In (B), the whole placental images were obtained by a Pannoramic MIDI scanner (3DHISTECH; Hungary) and analyzed using Case Viewer software. Scale bar, 500 μm. (C) Local enlargement of the placental labyrinth, obtained by a confocal LSM980 microscope and analyzed using ZEN 2.6 software. CK17/19 is used to stain trophoblast cells. Scale bar, 20 μm. Representative images from at least three placentas per genotype from at least two litters are shown. (D) GSEA of genes expressed in the placentas of poly(I:C)-treated mice compared to those expressed in the placentas of WT mice (saline-treated), showing inhibition of vasculogenesis pathway. (E) Placentas from poly(I:C)- or saline-treated pregnant WT or Rsad2^−/−mice were fixed in PFA. Paraffin-embedded sections were then stained for CD31 (used to stain blood vessel) with DAB. Representative blood vessel images in labyrinth zone from three placentas of three pregnant mice per genotype and per treatment are shown. Scale bar, 50 μm. (F) Quantification of the labyrinth zone area occupied by blood vessels, visualized using CD31 staining as described in (E) and analyzed using Case Viewer software, n = 240. Error bars indicate SEM, and statistical analyses were performed using the unpaired t test. For measuring vascular space, a researcher was blinded to the samples and asked to quantify the area within ∼10 blood vessels from each image irrespective of the orientation of the vessel cross-section. Blood vessels were defined using CD31 staining and were quantified from at least eight different fields of view from three different placentas for each group. (G and H) GSEA of genes expressed in the placentas of Rsad2^−/−mice compared to those expressed in the placentas of WT mice (poly[I:C]-treated), showing the enrichment of genes associated with the vasculogenesis (G) and erythrocyte development (H) pathways. (I) Comparison of FPKM values of Aplnr (Apelin receptor), Kdr (VEGF receptor 2), Tie1 (Tie1, angiopoietin receptor 1), Tek (Tie2, angiopoietin receptor 2), Apln (Aplin), and Apela (Elabela) in mouse placentas, quantified using RNA-seq; the unpaired t test was used for statistical analyses. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001; ns, not significant. To verify this notion, IHC staining of placenta tissue sections with an anti-CD31 antibody was conducted. With poly(I:C) treatment, the severe vascular injury was observed in WT mice, but not in Rsad2^−/−mice ([147]Figure 5E). Moreover, the size of the area occupied by blood vessels in the placenta was significantly decreased in poly(I:C)-treated WT mice compared with that in Rsad2^−/−mice, indicating that RSAD2 depletion alleviated placental vascular injury ([148]Figures 5E and 5F). We next analyzed transcriptional data from the placentas of WT and Rsad2^−/−mice following poly(I:C) treatment and found that vasculogenesis and the erythrocyte development pathway were significantly restored in Rsad2^−/−mice compared with the WT animals ([149]Figures 5G and 5H). Moreover, the genes encoding the apelin receptor (Aplnr), the VEGF receptor 2 (Kdr), and the angiopoietin receptors (Tie1 and Tie2), which mediate proangiogenic signaling in the placenta, were significantly downregulated in poly(I:C)-treated WT mice, but not in Rsad2^−/−mice ([150]Figure 5I). The adipokine apelin (encoded by Apln), a ligand of Aplnr, had an increased trend in the placenta of poly(I:C)-treated WT mice (relative to the PBS-treated WT controls), a result of a feedback effect. By contrast, the expression of Apln had a tendency to decrease in poly(I:C)-treated Rsad2^−/−mice (relative to poly(I:C)-treated WT mice) ([151]Figure 5I). The Apln-Aplnr axis is implicated in cardiovascular function and vasoconstriction.[152]^31 Apela, which encodes the peptide Elabela (an endogenous ligand of the Aplnr in placenta), is a circulating hormone secreted by the placenta to promote placental development and prevent preeclampsia.[153]^32 We found that the expression of Apela was higher in Rsad2^−/−mice ([154]Figure 5I). Our findings collectively showed that the high expression of Rsad2 in WT mice is associated with severe vessel injury in the placenta and the excessive accumulation of neutral lipids. RSAD2 depletion ameliorates APOs and lipid accumulation in pristane-induced pregnant mice To further demonstrate that Rsad2 is a pathogenic ISG in SLE pregnant model, we next used an improved SLE induction model. Pristane-induced lupus is a classical model of SLE with overactivation of IFN-I signaling.[155]^33 We found that the expression of Rsad2, Isg15, and Ifit1 in the decidua and placenta was upregulated in pregnant mice ([156]Figures S5A and S5B). To examine the links between the upregulation of Rsad2 and the pathogenesis of SLE pregnancy, Rsad2^−/− and WT pregnant mice were treated with pristane. Pristane challenge causes embryonic developmental abnormalities, especially the fetal brain and liver, and medium absorption (relative to the degree of embryo resorption in the poly[I:C] model) ([157]Figure S5C). We found that pristane treatment significantly reversed the phenotype of embryonic developmental abnormalities in Rsad2^−/−pregnant mice compared with WT pregnant mice at E12.5 ([158]Figures S5C and S5D). Compared with pristane-treated WT mice, embryonic developmental abnormalities and the embryo absorption rate significantly decreased in Rsad2^−/−pregnant mice ([159]Figure S5E). Moreover, the body length, head widths, and weight of live fetuses were significantly smaller in pristane-treated WT mice compared with saline-treated WT control mice ([160]Figure S5F). By contrast, the fetuses of the pristane-treated Rsad2^−/−mice were alleviated in size to those of the pristane-treated WT group ([161]Figure S5F). In conclusion, unlike the apparent APO phenotype in pristane-treated WT mice, the Rsad2^−/−mice with pristane-treatment significantly alleviated embryo resorption or abnormal development. The aforementioned results suggest that Rsad2, as a pathogenic ISG, induces adverse pregnancy outcomes in a pristane-induced SLE-like model. Consistent with the observations made in the poly(I:C) model, lipid accumulation-related genes were upregulated ([162]Figure S5G). Furthermore, following pristane treatment, unlike that LD accumulation was observed in the WT animals, the LD content of the decidua and labyrinthine zone in Rsad2^−/−mice was similar with saline-treated WT mice ([163]Figures S5H and S5I). In addition, vasculogenesis was significantly inhibited in the pristane-induced SLE model ([164]Figure S5J). Compared with that in WT mice with pristane inducement, the size of the area occupied by placental blood vessels was significantly increased in the Rsad2^−/− mice, indicating that RSAD2 depletion alleviated placental vascular injury ([165]Figures S5J and S5K). Taken together, our results show that Rsad2 is a pathogenic ISG, which causes adverse pregnancy outcomes by inducing placental lipid accumulation and vascular injury in a pristane-induced SLE-like model. RSAD2 triggers lipid accumulation in the spontaneous SLE pregnancy model and SLE patients To further explore whether the upregulation of Rsad2 caused fetal demise by promoting LD accumulation in the placenta, we used New Zealand White (NZW) and New Zealand Black (NZB) mice as spontaneous murine models of SLE. Among the NZW, NZB, C57BL/6J, and BALB/c pregnant mice, the worst pregnancy outcomes were observed in the NZW mice ([166]Figures 6A and [167]S6A). The lengths and widths of NZW live fetuses were significantly lower, and the fetal resorption rates were significantly higher than those of the NZB, C57BL/6J, and BALB/c mice ([168]Figures 6B and 6C). We found that the expression of Rsad2 in the uteruses of NZW mice was also higher than in those of the NZB, C57BL/6J, and BALB/c mice ([169]Figure 6D). To explore whether high Rsad2 expression promoted the accumulation of placental LDs in NZW mice, whole frozen placenta sections were stained with BODIPY 493/503. Indeed, the LD content of the decidua and the labyrinthine zone was significantly higher in the NZW mice compared with C57BL/6J animals ([170]Figures 6E and 6F). Oil red O staining was used to further characterize the effect of Rsad2 expression on placental LDs in NZW mice ([171]Figures 6G and 6H). Hematoxylin and eosin (H&E) and anti-CD31 antibody IHC staining of whole placenta tissue sections showed evidence of vessel wall thickening and a reduction in the number of blood vessels in the decidua and placenta of NZW mice ([172]Figures S6B and S6C). We next examined whether this phenomenon also occurred in pregnant patients with SLE. Indeed, we found that the LD content was also increased in the placental villi of patients with SLE using LD staining ([173]Figures 6I and 6J) and oil red O staining ([174]Figures 6K and 6L). Thus, the aforementioned results indicate that the upregulation of RSAD2 induced lipid accumulation in the placenta, leading to blood vessel injury and ultimately causing adverse pregnancy outcomes in spontaneous SLE murine models and patients with SLE. Figure 6. [175]Figure 6 [176]Open in a new tab Lipid accumulation at the maternal-fetal interface in a mouse model of spontaneous SLE and pregnant patients with SLE (A) One representative litter from each of the NZW, NZB, C57BL/6J, and BALB/c groups of mice, collected at gd12.5. Scale bar, 1 cm. Sires and dams of all groups for mating have the same genetic backgrounds. (B) Lengths (left) and widths (right) of live fetuses NZW (n = 4), NZB (n = 3), C57BL/6J (n = 4), and BALB/c (n = 5) mice were recorded and counted. (C) Pie charts showing the total number of fetuses analyzed and their developmental phenotypes (i.e., resorbed or halted development versus phenotypically normal), related to (A). (A), (B), and (C) are representative of at least two independent experiments. (D) The expression of Rsad2 and Isg15 in the uteruses of NZW, NZB, C57BL/6J, and BALB/c mice was detected using quantitative real-time PCR. Each dot represents a sample from one mouse. In (B) and (D), error bars indicate SEM and statistical analyses were performed using the unpaired Student’s t test. (E and G) (E) LD staining and (G) oil red O staining of decidua basalis (DB) and labyrinth zone (LZ) of NZW and C57BL/6J mice, collected at gd12.5. Images are representative of three independent experiments. Scale bar, 50 μm. (F) Quantification of LD staining of placental labyrinth using ZEN 2.6. (H) Quantification of oil red O staining using Image-Pro Plus 6.0. (I and K) (I) LD staining and (K) oil red O staining of the first trimester deciduas and chorionic villi from each of the healthy donors and patients with SLE. Images are representative of three heathy donors and three patients with SLE. Scale bar, 50 μm. (J) Quantification of LD staining of placental labyrinth using ZEN 2.6. (L) Quantification of oil red O staining using Image-Pro Plus 6.0. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001; ns, not significant. Virtual screening of a small-molecule compound targeting RSAD2 alleviates APOs Given that RSAD2 acts as pathogenic ISGs in SLE pregnancy, virtual screening of two commercialized small-molecule compound libraries targeting the mouse RSAD2 protein structure was conducted. L-chicoric acid (LCA) has the highest docking score, whose structural formula is shown in [177]Figure 7A. LCA can form 7 hydrogen bonding interactions and 3 π-π interactions with mouse RSAD2 protein (As a hydrogen bond donor, a phenolic hydroxyl group forms 1 hydrogen bonding interaction with LEU252, and another phenolic hydroxyl group forms 1 hydrogen bonding interaction with LYS299, both at a distance of 2.0 Å; a carboxyl group acts as a hydrogen bond acceptor and forms 1 hydrogen bonding interaction with SER124 at a distance of 2.3 Å, and the oxygen atom on another carboxyl group acts as a hydrogen bond acceptor with LYS220 and ASN222, LYS247, with distances of 2.4, 1.7, and 2.4 Å, respectively, and ASN222 forms one hydrogen bond with the carboxyl oxygen atom on the main chain, with a distance of 2.8 Å) ([178]Figure 7B). To investigate whether LCA has a therapeutic effect by targeting pathogenic RSAD2, the compound was injected to pregnant mice after poly(I:C) treatment. The results showed that pregnancy outcome was significantly alleviated in the LCA-treated mice compared to the vehicle-treated group ([179]Figures 7C–7F), while the level of RSAD2 did not change in LCA-treated mice. ([180]Figures S7A and S7B). APOs were alleviated, and embryo resorption rate was significantly reduced after LCA treatment ([181]Figures 7C and 7E). Individual fetuses were evaluated morphologically for appearance and size by taking photographs with a stereoscope and measuring the crown-rump length, the width of the fetal head, and the body weight of live fetuses with a vernier caliper and an analytical balance. We found that the size of fetuses (especially fetal brain and liver) was more closely resembling normal healthy embryonic size in LCA treatment mice compared to the vehicle group ([182]Figures 7D and 7F). LCA treatment significantly restored the postnatal survival rate and reversed the weight loss in offspring mice from poly(I:C)-treated dams, compared to the vehicle control ([183]Figures S7C and S7D). The aforementioned results demonstrate that LCA treatment targeting RSAD2 can alleviate adverse pregnancy outcomes in poly(I:C)-challenged mice. While LCA alleviates APOs, lipid accumulation in maternal decidual and placenta labyrinthine zone also reduced, especially the large size of LDs ([184]Figure 7G). Both H&E staining and immunohistochemistry of CD31 labeling of vascular endothelial cells of the placenta showed a significant improvement of vascular injury after LCA treatment compared with vehicle-treated mice ([185]Figures S7E and S7F). To examine whether LCA also has a therapeutic effect by targeting pathogenic RSAD2 in pristane-induced SLE-like model, the compound was injected to pregnant mice after pristane challenge. Consistent with previous results, LCA did not directly affect the expression level of RSAD2 ([186]Figures S7G and S7H), while the pregnancy outcome was alleviated in the LCA-treated mice compared to the vehicle-treated group ([187]Figures 7H–7K). Compared with the small size of individual fetuses observed in the vehicle group, the live fetuses had healthier morphology in LCA treatment mice ([188]Figure 7I). Measurement of the length, width, and body weight of live fetuses ([189]Figure 7K), and the weight of offspring mice ([190]Figure S7J), also illustrates the same conclusion. More to the point, LCA treatment significantly attenuated the rate of embryo absorption and abnormal development during pregnancy ([191]Figure 7J) and the survival rate of offspring ([192]Figure S7I). Moreover, the accumulation of LDs of placenta sections from LCA-treated dams was lower than that from the vehicle group ([193]Figure 7L). Both H&E staining and IHC of placental blood vessel suggested a significant improvement of vascular damage after LCA treatment ([194]Figures S7K and S7L). Surprisingly, LCA treatment reduced the proportion of embryonic resorption in NZW mice ([195]Figure S7M), a model of spontaneous SLE that has previously been shown to have serious fertility problems ([196]Figure 6C), although no significant improvement in embryonic size ([197]Figure S7N). In conclusion, a small-molecule compound LCA targeting pathogenic RSAD2 alleviates APOs in poly(I:C)-challenged mice, pristane-induced SLE-like model, and NZW mice, which provides new target and therapeutic ideas for the clinical improvement of adverse pregnancy outcomes in women with SLE and other pregnancies with abnormal activated IFN-I signal. Figure 7. [198]Figure 7 [199]Open in a new tab Virtual screening of a small-molecule compound targeting RSAD2 alleviates APOs (A) Structural formulae of LCA compound. (B) 3D diagram (left) of the binding pattern of LCA and RSAD2 and the detailed binding patterns were further analyzed (right). LCA was colored with rose stick. C skeleton of mouse RSAD2 protein is shown in green, N atoms are shown in blue, O atoms are shown in bright red, and H atoms are shown in white. (C–F) Poly(I:C) was injected at E10.5. 10 mg/kg LCA and vehicle were intraperitoneally injected at E10.5 and E11.5. (C) Representative litter from vehicle-treated and LCA-treated groups of mice, collected at gd12.5. Scale bar, 1 cm. (D) One representative litter from each of the indicated treatment groups collected at gd12.5 is shown. Scale bar, 1 mm. (E) Pie charts showing the total number of fetuses analyzed and their developmental phenotypes (i.e., resorbed or halted development versus phenotypically normal), related to (C). Saline-vehicle (n = 4), saline-LCA (n = 4), poly(I:C)-vehicle (n = 4), and poly(I:C)-LCA (n = 4) were recorded. (F) Lengths (left), widths (middle), and weights (right) of live fetuses from indicated each group were recorded and measured. (C)–(F) are representative of at least two independent experiments. (G) Oil red O staining of decidua basalis (DB) and labyrinth zone (LZ) of LCA or vehicle-treated mice with same poly(I:C) treatment, which is harvested at E12.5. Representative image of placentas from at least three dams. Scale bar, 50 μm. (H–K) 10 mg/kg LCA was injected at E10.5 after induced with 500 μL pristane or same volume saline, and vehicle was injected to control mice at the same time. (H) Representative litter from vehicle-treated and LCA-treated groups of mice, collected at gd12.5. Scale bar, 1 cm. (I) One representative litter from each of the indicated treatment groups collected at gd12.5 is shown. Scale bar, 1 mm. (J) Pie charts showing the total number of fetuses analyzed and their developmental phenotypes (i.e., resorbed or halted development versus phenotypically normal), related to (H). 6–7 dams per group enrolled. (K) Lengths (left), widths (middle), and weights (right) of live fetuses from indicated each group were recorded and measured. Saline-vehicle (n = 3), saline-LCA (n = 3), pristane-vehicle (n = 5), and pristane-LCA (n = 7) were recorded. (H)–(K) are representative of two independent experiments. (L) Oil red O staining of DB and LZ of LCA or vehicle-treated mice with pristane inducement, collected at gd12.5. Images are representative of three independent experiments. Scale bar, 50 μm. In (F) and (K), error bars indicate SEM, and statistical analyses were performed using the unpaired Student’s t test. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001; ns, not significant. Discussion Our work examined SLE pregnancy as a model and revealed that RSAD2 acts as a pathogenic ISG. Unlike the protective ISGs, RSAD2 promotes excessive DAG-lipid accumulation at the maternal-fetal interface, causing placental vascular damage and poor pregnancy outcomes. In addition, LCA, as a new inhibitor targeting RSAD2 by virtual screening, significantly alleviated APOs and lipid accumulation in three pregnant mice model with dysregulated IFN-I signaling. Thus, RSAD2 may represent a promising target in the treatment of adverse pregnancy outcomes caused by an abnormally activated IFN-I response. Anifrolumab, a human monoclonal antibody to IFN-I receptor subunit 1, has been investigated for the treatment of SLE.[200]^34 The clinical trial showed that patients who received anifrolumab experienced a positive outcome. However, there was a higher incidence of herpes zoster and bronchitis.[201]^35 The trial assessing the long-term safety of anifrolumab revealed higher exposure-adjusted event rates of COVID-related adverse events, including asymptomatic infections, compared to the placebo.[202]^36 It is suggested that distinguishing between pathogenic and protective ISGs offers the potential for the precise treatment of SLE or SLE-related pregnant disorders. Several studies have demonstrated that the abnormal activation of placental IFN-I signaling leads to adverse pregnancy outcomes during viral infection.[203]^2^,[204]^3^,[205]^15 A previous study found that IFITMs inhibited placental syncytial trophoblast fusion, leading to embryonic loss.[206]^2 This provided a possible explanation for how IFN-I and its downstream pathogenic ISGs mediated placental dysfunction in a mouse pregnancy model stimulated by the IFN-I inducer poly(I:C). In our study, we found that RSAD2 caused placental damage by perturbing lipid metabolism at the maternal-fetal interface and led to placental vascular injury in SLE. In addition, poly(I:C) injections at different gestational stages resulted in different changes in pregnancy outcomes. Existing literature shows that starting poly(I:C) injections at E7.5—before placental formation—leads to nearly complete embryo absorption.[207]^2 Injection of poly(I:C) typically results in a low probability of embryonic loss after E12.5.[208]^37 Based on the adverse pregnancy outcomes observed in clinical cases of SLE and SLE mouse models, we opted to administer poly(I:C) at E10.5 to better simulate adverse outcomes such as stillbirth and intrauterine growth restriction. RSAD2 encodes an IFN-inducible protein, RSAD2 (also named viperin, CIG5, CIG33), which is a member of the radical S-adenosyl-L-methionine superfamily of enzymes. RSAD2 inhibits the replication of multiple viruses by producing the ribonucleotide 3′-deoxy-3′,4′-didehydro-cytidine triphosphate.[209]^38^,[210]^39^,[211]^40 However, RSAD2 facilitates the infectious process by disrupting cellular lipid metabolism during infection with human cytomegalovirus. Meanwhile, RSAD2 was relocalized from the endoplasmic reticulum to the mitochondria.[212]^41^,[213]^42 RSAD2 is considered to be a key diagnostic ISG in SLE and may contribute to the personalized targeted treatment of this disease.[214]^43 Moreover, RSAD2 is significantly upregulated in atherosclerosis, where its expression is regulated by proinflammatory agents (i.e., by lipopolysaccharide, cytomegalovirus, and IFN-γ but not by tumor necrosis factor alpha or IL-1β).[215]^44 RSAD2 is expressed in the atherosclerotic lesions of humans and Apoe^−/−mice but not in normal arteries.[216]^44 RSAD2 expression is also observed in various types of cancer, where RSAD2-mediated metabolic alterations drive the progression of tumors.[217]^45 The aforementioned studies suggest that RSAD2 is a pathogenic ISG in pregnant patients with SLE and others with an overactive IFN-I response. In this study, we reported a novel role for RSAD2 in the regulation of cellular metabolism at the maternal-fetal interface in individuals with a disease that affected pregnancy outcomes. We found that RSAD2 may lead to abnormal embryonic dysplasia by inducing the accumulation of sn-1,2-DAGs in the placenta. Through virtual screening, we found one small-molecule compound targeting RSAD2. As a dicaffeoyltartaric acid, LCA is a potent, selective, and reversible HIV-1 integrase inhibitor.[218]^46 Chicoric acid is isolated and purified from plant and vegetables, which is reported to possess antioxidant and anti-inflammatory activities.[219]^47 Given that LCA has effects on alleviating adverse pregnancy outcomes in treating the SLE mice models and its natural product properties, small-molecule compound therapy targeting RSAD2 has potential clinical applications. In addition, elevated RSAD2 has been observed in atherosclerotic plaques, where it is associated with lipid accumulation and foam cell formation, exacerbating the disease.[220]^48 This is similar to the phenomenon observed in the placental labyrinth zone of our SLE pregnancy model, expanding the potential role of RSAD2 as a target in vascular inflammatory diseases and the therapeutic value of LCA. Interestingly, we found that IFN-I stimulation upregulated APOD in the decidua ([221]Figure S4H), while its receptor-encoding genes LRP1 and SR-B1 were highly expressed in the placental region (data not showed). The APOD-LRP1/SR-B1 signaling axis may mediate lipid transfer from the decidua to the placenta. Our previous research found that decidual NK cells used the APOD-LRP1 signaling axis to deliver lipids to trophoblasts, which helped trophoblasts resist apoptosis (induced by intracellular bacterial infection) and promoted their survival.[222]^49 In an IFN-I-rich environment, the upregulation of APOD expression in DSCs could further transport lipids from the decidual tissue to the placental trophoblast layer, leading to the abnormally excessive accumulation of LDs in the placenta. In patients with atherosclerosis, macrophages induced vascular injury by phagocytosing lipids.[223]^50 Whether placental macrophages are responsible for the induction of placental injury via disrupting lipid metabolism deserves further investigation, and whether RSAD2 contributes to the proatherogenic effects of IFN-α in SLE also deserves further investigation. Ultimately, our work provides new insights into how targeting pathogenic ISGs may benefit pregnant patients with diseases associated with dysregulated IFN-I signaling. Limitations of the study Although we have shown that RSAD2 is mainly expressed in structural cell subpopulations using purified cells, we used full rather than conditional knockout mice based on the fact that RSAD2 is broadly expressed at the maternal-fetal interface. Our results cannot rule out the possibility that RSAD2 plays a role at other sites in SLE, such as the spleen and kidney. In addition, RSAD2 plays an important antiviral infection role by inhibiting viral replication.[224]^39 Thus, RSAD2 deficiency may not be beneficial in some mouse models of viral (i.e., ZIKA) infection-induced APOs. The factors contributing to adverse pregnancy outcomes in patients with SLE are complex, including antinuclear antibodies, complement proteins, inflammatory cytokines, and vascular factors.[225]^51 Our study explains the molecular basis of risky pregnancies associated with an overactive IFN-I response in the SLE model. Resource availability Lead contact Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Haiming Wei (ustcwhm@ustc.edu.cn). Materials availability Plasmids generated in this study are available from the [226]lead contact without restriction. Data and code availability * • RNA-seq data files, quasi-targeted metabolomics, and widely targeted quantitative lipidomics are available in the GSA or OMIX database repository of the National Genomics Data Center, China National Center for Bioinformation. These data are publicly available as of the date of publication. Accession numbers are listed in the [227]key resources table. * • This study did not report new original code. * • Any additional information required to reanalyze the data reported in this paper is available from the [228]lead contact upon request. Acknowledgments