Endothelial erα promotes glucose tolerance by enhancing endothelial insulin transport to skeletal muscle

Endothelial erα promotes glucose tolerance by enhancing endothelial insulin transport to skeletal muscle

Play all audios:

Loading...

ABSTRACT The estrogen receptor (ER) designated ERα has actions in many cell and tissue types that impact glucose homeostasis. It is unknown if these include mechanisms in endothelial cells,


which have the potential to influence relative obesity, and processes in adipose tissue and skeletal muscle that impact glucose control. Here we show that independent of impact on events in


adipose tissue, endothelial ERα promotes glucose tolerance by enhancing endothelial insulin transport to skeletal muscle. Endothelial ERα-deficient male mice are glucose intolerant and


insulin resistant, and in females the antidiabetogenic actions of estradiol (E2) are absent. The glucose dysregulation is due to impaired skeletal muscle glucose disposal that results from


attenuated muscle insulin delivery. Endothelial ERα activation stimulates insulin transcytosis by skeletal muscle microvascular endothelial cells. Mechanistically this involves nuclear


ERα-dependent upregulation of vesicular trafficking regulator sorting nexin 5 (SNX5) expression, and PI3 kinase activation that drives plasma membrane recruitment of SNX5. Thus, coupled


nuclear and non-nuclear actions of ERα promote endothelial insulin transport to skeletal muscle to foster normal glucose homeostasis. SIMILAR CONTENT BEING VIEWED BY OTHERS DEFICIENCY OF


ENDOTHELIAL SIRTUIN1 IN MICE STIMULATES SKELETAL MUSCLE INSULIN SENSITIVITY BY MODIFYING THE SECRETOME Article Open access 11 September 2023 PARACRINE ROLE OF ENDOTHELIAL IGF-1 RECEPTOR IN


DEPOT-SPECIFIC ADIPOSE TISSUE ADAPTATION IN MALE MICE Article Open access 02 January 2025 LOSS OF BONE MORPHOGENETIC PROTEIN-BINDING ENDOTHELIAL REGULATOR CAUSES INSULIN RESISTANCE Article


Open access 26 March 2021 INTRODUCTION In addition to their classic roles in reproduction, estrogens have a potent impact on glucose homeostasis1,2. In female rodents and primates,


ovariectomy leads to glucose intolerance and insulin resistance particularly in the setting of diet-induced obesity, and these effects are reversed by estrogens2,3,4,5. Estrogen receptors


(ER) are members of the steroid hormone receptor superfamily that traditionally serve as transcription factors6, and the ER designated ERα modulates multiple aspects of glucose


regulation2,3,4,5. The known relevant sites of action include the CNS, liver, skeletal muscle myocytes, pancreas, macrophages and adipose tissue7,8,9,10,11,12. In women, surgical or natural


menopause increases the age-adjusted odds ratio of developing diabetes to 1.59 (CI 1.07-2.37) compared to the rate in premenopausal women13. In postmenopausal women, changes in glucose


homeostasis also occur14, and hormone replacement therapy (HRT) with estrogens lowers the risk of developing diabetes15. In men, individuals with disruptive mutations in the ERα gene or the


gene encoding aromatase, which produces estrogens by aromatizing androgens, have decreased glucose tolerance, and in the latter category 17β-estradiol (E2) administration improves glucose


tolerance16,17,18. In parallel, male aromatase-/- mice have insulin resistance that is reversed by E219. Thus, estrogens and ERα have important impact on glucose homeostasis in both males


and females. Whereas the metabolic effects of estrogens partnering with ERα are well known in many cell types and tissues including adipocytes and skeletal muscle myocytes, the participation


of ERα in endothelium in glucose control is unknown. Interestingly, in addition to predictably regulating endothelial gene expression, in endothelial cells there is a plasma


membrane-associated subpopulation of ERα which uniquely mediates numerous non-nuclear processes, including E2 activation of endothelial NO synthase (eNOS)20,21,22,23. Mechanisms in


endothelial cells influence both relative obesity and other processes in adipose that impact glucose homeostasis. Effective vascularization is essential to the healthy expansion of adipose


tissue24, the endothelium governs adipose tissue inflammation and thereby influences adipokine and cytokine production25, and endothelial ERα actions are both proangiogenic and


anti-inflammatory26,27,28. Based on these rationales, in the current work, experiments were designed in mice to determine if and how ERα in endothelium influences glucose homeostasis. They


revealed that endothelial ERα excision in both male and female mice leads to glucose intolerance and insulin resistance, and surprisingly this was not due to changes in adiposity or


processes in adipose that influence glucose control. Efforts then turned to the interrogation of endothelial mechanisms in other tissues involved in glucose control, and they uncovered a


defect in skeletal muscle glucose disposal related to an impairment in muscle insulin delivery. Further work ultimately identified coupled nuclear and non-nuclear actions of ERα which


promote endothelial insulin transport by upregulating expression of the vesicular trafficking regulator sorting nexin 5 (SNX5) and by activating PI3 kinase to drive plasma membrane


recruitment of SNX5. These findings reveal a mode of action of estrogens and ERα that may represent a set of targets to leverage to improve glucose tolerance and insulin sensitivity. RESULTS


ENDOTHELIAL ERΑ PROMOTES GLUCOSE CONTROL BY ENHANCING MUSCLE INSULIN DELIVERY To begin determining how ERα in endothelium impact glucose homeostasis, fasting glucose and glucose tolerance


tests (GTT) were performed in control floxed ERα mice (ERαfl/fl) and mice selectively deficient in the receptor in endothelial cells (ERαΔEC, Supplementary Fig. 1a, b)29. Male ERαΔEC mice


displayed fasting hyperglycemia and abnormal glucose tolerance tests (GTT), and they also had elevated HOMA-IR, and decreased glucose infusion rates (GIR) during a hyperinsulinemic,


euglycemic clamp (Fig. 1a–d). In HFD-fed, ovariectomized females whereas E2 caused a dramatic normalization of GTT in ERαfl/fl controls, the capacity of E2 to prevent HFD-induced glucose


intolerance was completely absent in ERαΔEC (Fig. 1e). This finding is consistent with a prior study limited to GTTs that demonstrated glucose intolerance in HFD-fed intact female mice


deficient in endothelial ERα30. Insulin tolerance tests (ITT) further revealed that the previously recognized capacity for E2 to improve insulin sensitivity is entirely absent in ERαΔEC


females (Fig. 1f). In light of numerous mechanisms by which endothelial cells in adipose tissue influence relative adiposity and glucose control, body composition was next evaluated.


Standard chow-fed male ERαfl/fl and ERαΔEC mice had similar body weights, percent body fat and lean body mass (Supplementary Fig. 1c–e), and subcutaneous (inguinal) and visceral (gonadal)


white adipose tissue (WAT) depots were similar in size (Supplementary Fig. 1f, g). In ovariectomized high-fat diet (HFD)-fed females, there was a partial blunting of E2-related body weight


loss in ERαΔEC, but E2-induced decreases in fat mass and increases in lean body mass were unaffected by endothelial ERα deficiency (Supplementary Fig. 1h–j). Thus, endothelial ERα does not


influence adiposity in male mice or mediate the well-recognized protection from adiposity by E2 in female mice5. The basis by which endothelial cell ERα influences glucose homeostasis was


then further pursued in male mice. Recognizing that adipose tissue vascularity impacts insulin sensitivity24, transcript abundance for the endothelial markers Flk1 and PECAM1 was evaluated


in subcutaneous and visceral WAT, and it was similar in ERαfl/fl and ERαΔEC mice (Supplementary Fig. 2a–d). Since the endothelium within fat depots governs the degree of inflammation25,


inflammatory cell abundance and cytokine expression were evaluated (Supplementary Fig. 2e–l), and levels of the macrophage marker F4/80, the leukocyte marker CD11, and IL-6 and TNFα


transcripts were similar in the WAT depots of the two genotype groups. As such, endothelial ERα does not influence adipose tissue vascularity or inflammation, and the impact of endothelial


ERα on glucose homeostasis entails processes other than those in adipose microvasculature that impact insulin sensitivity24,25. Since the endothelium in the pancreas influences insulin


secretion from beta cells31, pancreatic insulin production was evaluated, and increases in plasma insulin levels in response to an acute glucose load were similar in ERαfl/fl versus ERαΔEC


mice (Fig. 1g). Despite this finding, the insulin response of the pancreas may not be fully preserved in the ERαΔEC mice because they display relative fasting hyperglycemia that is not


compensated by an increase in insulin secretion. Noting that processes in hepatic endothelium influence insulin action in the liver32, possible changes in hepatic insulin sensitivity were


studied using pyruvate tolerance tests, and they were similar in ERαΔEC and ERαfl/fl mice (Fig. 1h). Thus, ERα in endothelium does not impact hepatic insulin sensitivity, and if there is an


impact on pancreatic insulin secretion, it is quite modest. Next, we interrogated processes in the skeletal muscle, where up to 80% of whole-body glucose disposal occurs in both humans and


rodents33,34. Glucose uptake in soleus and gastrocnemius was decreased by 33% in ERαΔEC males (Fig. 1i), and in HFD-fed ovariectomized females, whereas E2 caused a 47% increase in glucose


uptake in ERαfl/fl controls, the increase in uptake with E2 was completely absent in ERαΔEC (Fig. 1j). Insulin action in skeletal muscle was then queried in males by quantifying the


activation of Akt kinase35, and compared to ERαfl/fl controls, Akt Ser473 phosphorylation in response to insulin was attenuated in ERαΔEC employing either total Akt or GAPDH as the


denominator (Fig. 1k–m, Supplementary Fig. 3a, b). Recognizing that mechanisms in the endothelium are critically involved in insulin delivery to the skeletal muscle myocytes36,37,38,39,


muscle insulin delivery was tested. Whereas there was a 2.7- to 3-fold increase in skeletal muscle insulin content 5 min after bovine insulin injection in ERαfl/fl controls, the increase was


blunted by 60 to 64% in ERαΔEC mice (Fig. 1n, Supplementary Fig. 3c). To determine if endothelial ERα impacts a process specifically in endothelial cells and not in myocytes to alter muscle


glucose uptake, we evaluated insulin-induced glucose uptake in isolated skeletal muscles ex vivo (Fig. 1o, p). In both soleus and EDL insulin-induced glucose uptake ex vivo was identical in


muscle from ERαfl/fl and ERαΔEC mice. This indicates that there are no changes in skeletal muscle myocyte processes regulating glucose homeostasis with endothelial ERα silencing independent


of the effects on endothelial insulin transport. Thus, through endothelium-based mechanisms, ERα in endothelium supports skeletal muscle glucose disposal by promoting insulin delivery to


muscle. ENDOTHELIAL ERΑ PROMOTES ENDOTHELIAL INSULIN TRANSPORT VIA BOTH NUCLEAR AND NON-NUCLEAR MECHANISMS How endothelial ERα positively influences skeletal muscle insulin delivery was then


determined. Since muscle insulin delivery and resulting glucose disposal are enhanced by insulin stimulation of skeletal muscle microvascular recruitment and blood flow40,41,42, the impact


of endothelial ERα on muscle microvascular functional responses to insulin was evaluated. This was accomplished using contrast-enhanced ultrasound (CEUS) with a lipid-shelled


perfluorocarbon-based microbubble (MB) contrast agent to image the muscle microvasculature before and during a hyperinsulinemic-euglycemic clamp. A representative still image of a CEUS


region-of-interest (ROI) is shown in Fig. 2a, and a sample time-intensity curve is in Fig. 2b. Analysis of the curve yields parametric measures of capillary blood volume (CBV: peak


intensity, _Ipk_), microvascular blood flow (MBF: time to peak intensity, _Tpk_; and wash-in rate, _WIR_), and both CBV and MBF (area under the curve, _AUC_; and wash-out rate,


_WOR_)43,44,45,46,47,48. Supplementary Movies 1 and 2 show real-time CEUS imaging of MB in the skeletal muscle microvasculature of an ERαfl/fl control mouse at baseline and during a clamp,


respectively. The greater MB signal seen in the clamp movie, particularly early in the movie, reveals how insulin increases capillary recruitment and blood flow in the skeletal muscle. In


the ERαfl/fl control mice whereas there was no change in Tpk in response to insulin (Fig. 2c), Ipk, WIR, AUC and WOR increased (Fig. 2d–g), indicative of insulin-induced increases in CBV and


MBF. Equal increases in Ipk, WIR, AUC and WOR occurred in ERαΔEC, revealing that the promotion of muscle insulin delivery by endothelial ERα is not related to effects on insulin-induced


microvascular recruitment and blood flow. Since changes in intracellular vesicles have been previously observed in skeletal muscle capillary endothelium in mice with obesity-related insulin


resistance49, such endothelial vesicles were then compared in ERαfl/fl and ERαΔEC mice (Supplementary Fig. 4a). Between genotype groups, the total number of vesicles per unit area of


endothelium was similar, as was the number of vesicles associated with the luminal plasma membrane or the abluminal plasma membrane, or localized intracellularly (Supplementary Fig. 4b, c).


Interestingly, independent of genotype group, the number of vesicles associated with the abluminal plasma membrane was greater than those on the luminal plasma membrane or those localized


intracellularly. Parallel findings were obtained regarding vesicle area relative to the endothelial area (Supplementary Fig. 4d, e). In addition, vesicle size was similar in ERαfl/fl and


ERαΔEC mice, and it was comparable in the three categories of vesicles (Supplementary Fig. 4f, g). Thus, changes in the frequency or size of capillary endothelial vesicles do not explain the


observed differences in skeletal muscle insulin delivery in ERαfl/fl and ERαΔEC mice. Along with insulin-related changes in CBV and MBF, insulin delivery to skeletal muscle is controlled by


processes governing its transcytosis across the endothelial monolayer, and there is evidence that the latter is rate-limiting for peripheral insulin action36,37,38,39. We therefore


determined if ERα influences insulin transport by endothelial cells, first interrogating insulin uptake, the initial step in transcytosis, in human aortic endothelial cells (HAEC). As


visualized in Fig. 3a and quantified in Fig. 3b, E2 caused a dramatic increase in the uptake of FITC-conjugated insulin, and the response was negated by the selective ERα antagonist


methyl-piperidino-pyrazone (MPP). Knowing that PI3 kinase mediates many of the processes governed by the subpopulation of non-nuclear ERα in endothelial cells50, the effect of the PI3 kinase


inhibitor LY294002 was tested, and it too fully prevented the stimulation of insulin uptake by E2 (Fig. 3a, b). Since the in vivo observations pertain to skeletal muscle microvascular


endothelium, we then studied human skeletal muscle microvascular endothelial cells (HSMEC), and found that E2 causes an ERα- and PI3 kinase-dependent increase in insulin uptake in HSMEC


(Fig. 3c). Endothelial insulin transcytosis was then evaluated, and E2 caused a more than 10-fold ERα- and PI3 kinase-dependent increase in insulin transcytosis in HAEC (Fig. 3d, e).


Importantly, parallel findings for insulin transcytosis were made in HSMEC (Fig. 3f). In the transwell assays transendothelial electric resistance (TEER) was in the range of 350-450 Ωcm2 and


similar in the various study groups within each experiment (Supplemental Fig. 5a–c), indicating that comparable monolayer barriers were present, and strengthening the evidence that the


insulin transport being interrogated is transcellular51. In primary mouse skeletal muscle endothelial cells, in which studies of insulin uptake are feasible, E2 caused a 3.2-fold increase in


uptake in endothelium from ERαfl/fl mice which was prevented by PI3 kinase inhibition, and uptake was not stimulated by E2 in endothelium from ERαΔEC mice (Fig. 3g). These findings


strengthen the evidence that ERα and PI3 kinase are required, and they help link the in vivo and cell culture observations. Demonstrating a requirement for PI3 kinase activation, we then


determined if non-nuclear actions of endothelial ERα are sufficient to promote endothelial insulin transcytosis employing the estrogen dendrimer conjugate (EDC), which selectively activates


non-nuclear ER26. In contrast to E2, EDC did not stimulate insulin transcytosis in either HAEC or HSMEC (Fig. 3h, i, Supplementary Fig. 5d, e). This observation is consistent with our prior


demonstration in ovariectomized, HFD-fed female mice that EDC does not afford the protection from insulin resistance observed with E2 treatment52. Thus, both the initial step of insulin


uptake and the capacity of endothelial cells to transcytose insulin are potently stimulated by endothelial ERα activation, and these processes require both nuclear and non-nuclear actions of


the receptor, with the latter entailing PI3 kinase activation. INTERROGATING THE ENDOTHELIAL ERΑ INTERACTOME AND TRANSCRIPTOME To further investigate the basis by which ERα in endothelial


cells promotes insulin transport, we leveraged the observation that non-nuclear and nuclear actions of the receptor are required, and queried both sets of processes using non-biased


approaches. Since non-nuclear ERα function in endothelial cells requires dynamic receptor interaction with other proteins50, we employed immunoprecipitation (IP) and liquid


chromatography/tandem mass spectrometry (LC/MS-MS) to interrogate the ERα interactome in endothelial cells and how it is influenced by E2 (Fig. 4a, Supplementary Fig 6a). E2 treatment for 30


 min caused both the recruitment and the dissociation of numerous proteins from the receptor. Supplementary Table 1 lists the 449 proteins recruited to ERα and the 214 proteins dissociated


from the receptor upon E2 liganding. Pathway analysis using biological process terms from Gene Ontology analysis53 revealed that a number of the proteins disassociated from the receptor


mediate processes in leukocytes, whereas a number of the proteins recruited to ERα are involved in cell export (Supplementary Fig. 6b, c). The findings regarding the ERα interactome in


endothelial cells were compared to 3 available reports for ERα interacting proteins in MCF-7 breast cancer cells (Supplementary Table 2). They interrogated the ERα interactomes in MCF-7


cells involving extra-nuclear proteins54,55 or nuclear proteins56. When the combined findings in these reports are compared to the present observations for proteins disassociated from ERα in


response to E2 in endothelial cells, the commonality includes proteins involved in membrane localization (Supplementary Fig. 6d, e); for proteins recruited to the receptor with E2 treatment


in endothelial cells, the common proteins are involved in cytoskeletal organization (Supplementary Fig. 6f, g). To query the nuclear actions of the E2/ERα tandem that promote microvascular


endothelial insulin transport in skeletal muscle, we interrogated the translatome in mouse skeletal muscle microvascular endothelial cells in vivo. Translating ribosome affinity purification


(TRAP) followed by RNA-seq (TRAPseq) was performed in mice expressing a GFP-tagged version of the ribosomal protein L10a selectively in endothelial cells (GFP-L10aEC). To establish the


approach, skeletal muscle (gastrocnemius and soleus) was harvested from the GFP-L10aEC mice, total RNA was isolated from the muscle, and TRAP RNA was purified from the muscle endothelium by


anti-GFP immunoprecipitation. Q-RT-PCR revealed that whereas RNAs for the myocyte-specific genes myogenin (MyoG), myosin D1 (MyoD1) and alpha-smooth muscle actin (α-SMA) were enriched in the


whole tissue and not in the TRAP RNA (Fig. 4b), RNAs for the endothelium-specific genes roundabout (Robo), PECAM-1, and VECad were highly enriched in the TRAP RNA (Fig. 4c). E2 modulation


of gene translation in the skeletal muscle microvascular endothelium was then interrogated by TRAPseq in the skeletal muscle of ovariectomized female GFP-L10aEC mice treated with E2 for 4


weeks (Fig. 4d). As is shown in the volcano plot in Fig. 4e, the translation of a large number of genes was downregulated or upregulated in the skeletal muscle microvascular endothelium in


response to E2 treatment. Genes demonstrating upregulated and downregulated translation are listed in Supplementary Table 3, along with their human homologs. TRAPseq on muscle microvascular


endothelium will also be useful in other paradigms in mice in which muscle endothelial insulin transport is altered, such as in the setting of insulin resistance caused by diet-induced


obesity48. Next, we pursued the concept that genes upregulated by E2 whose gene products are recruited to E2-liganded ERα in endothelial cells may reveal those involved in the promotion of


endothelial insulin transport, and merged the findings by IP-LC/MS-MS and TRAPseq. The overlap of E2-downregulated ERα-interacting proteins and transcripts identified 27 genes (Fig. 4f, h


and Table 1), some of which are involved in mRNA processing. Conversely, with E2 treatment there were 23 proteins recruited to ERα whose gene translation was also increased by E2 (Fig. 4g,


i, and Table 1). Interestingly these included a number involved in metabolic processes. ERA PROMOTES ENDOTHELIAL INSULIN TRANSPORT VIA COUPLED NUCLEAR AND NON-NUCLEAR ACTIONS ON SNX5 As


highlighted in Fig. 4g, one of the 23 endothelial genes with increased translation by TRAPseq and increased gene product recruitment to ERα by LC/MS-MS in response to E2 was sorting nexin 5


(SNX5). SNX5 is a member of the SNX protein family of cytoplasmic and membrane-associated proteins that mediate vesicular trafficking of plasma membrane proteins57,58,59. Immunoblotting of


the anti-ERα coimmunoprecipitated proteins from HAEC subjected to LC/MS-MS displayed SNX5 following cell treatment with E2 (Fig. 5a). Independent co-immunoprecipitation studies demonstrated


parallel E2 promotion of ERα-SNX5 interaction in HAEC and HSMEC (Fig. 5b, c), and E2-induced upregulation of SNX5 expression was directly demonstrated in both HAEC and HSMEC (Fig. 5d, e). In


addition, E2 enhancement of SNX5 gene translation in vivo in the skeletal muscle microvascular endothelium was shown by QPCR on TRAP RNA (Fig. 5f). To determine if nuclear actions of ERα


mediate the upregulation of SNX5 expression by E2, experiments were performed in HAEC in which endogenous ERα was replaced with either wild-type ERα (WT) or two mutant forms of ERα with


altered nuclear function (Fig. 5g). Whereas reconstitution with WT ERα caused a return of SNX5 upregulation in response to E2, upregulation did not occur in cells harboring mutant ERα


lacking nuclear localization signals 2 and 3 (ERαΔ250-274) or the DNA binding domain (ERαΔ185-251)60,61. These findings are consistent with the observations made using the nonbiased


approaches, confirming that the activation of ERα increases both SNX5 expression through nuclear actions of the receptor and SNX5 interaction with the receptor in endothelial cells. SNX5 and


related SNX family members contain a phosphoinositide-binding Phox (or PX) domain which binds PIP3 to mediate SNX5 localization to membranes62. Recognizing that E2 liganding of ERα in


endothelial cells activates PI3 kinase50, and having found that E2 stimulation of endothelial cell insulin transport is PI3 kinase-dependent (Fig. 3b, c, e–g), we determined if PI3 kinase


participates in the modulation of SNX5 in endothelium by the E2-ERα tandem. LY294002 treatment prevented E2 promotion of ERα-SNX5 interaction in both HAEC and HSMEC (Fig. 5h, i), in response


to E2 both ERα and SNX5 were recruited to the plasma membrane, and the SNX5 trafficking required PI3 kinase (Fig. 5j–l). As such, the requirement for PI3 kinase stimulation in E2-related


enhancement of endothelial insulin transport likely reflects the role of the kinase in SNX5 recruitment to the plasma membrane and to ERα. In addition, RNAi was employed to determine if


there is a requirement for SNX5 in E2 stimulation of endothelial cell insulin transport in HAEC and HSMEC. Importantly, the knockdown of SNX5 did not alter insulin receptor or insulin


receptor substrate-1 expression in endothelial cells, as was previously observed in renal proximal tubule cells (Supplementary Fig. 7a–c, e–g)63. Since insulin receptor substrate-2 in


endothelial cells is critical to insulin transport64, its expression was also assessed and it was unchanged by SNX5 knockdown (Supplementary Fig. 7a, d, e, h). However, the loss of SNX5


fully attenuated E2-stimulated insulin uptake and transcytosis in both HAEC and HSMEC (Fig. 5m–p). Since insulin transcytosis requires its endocytosis, the combined findings likely indicate


that the primary process modulated by the coupling of ERα to SNX5 is insulin endocytosis. Finally, SNX5 reconstitution studies demonstrated that the loss of E2-stimulated insulin uptake with


SNX5 silencing is not due to off-target effects of SNX5 RNAi (Fig. 5q). Thus, SNX5 is a linchpin that couples ERα to endothelial cell insulin transport (Fig. 5r), providing the molecular


underpinnings of the first demonstration of a steroid hormone receptor promoting a transcytotic process. DISCUSSION Serving as both a transcription factor and an initiator of non-nuclear


signaling in response to estrogens20,65, it is well recognized that ERα in endothelial cells modulates angiogenesis and inflammation26,27,28. Since angiogenesis and inflammation greatly


influence mechanisms in adipose tissue and adipose impact on glucose homeostasis24,25, in our interrogation of endothelial ERα impact on metabolism, it was surprising to find a lack of


effect of endothelial ERα on adipose tissue. Instead, we discovered that endothelial ERα promotes glucose tolerance by enhancing insulin delivery to skeletal muscle and thereby increasing


glucose disposal. We further found that this is not related to impact on insulin-induced capillary recruitment or blood flow in muscle, or changes in skeletal muscle capillary endothelium


vesicle structure. Alternatively, the transcytosis of insulin across skeletal muscle endothelial cells is potently stimulated by E2 activation of endothelial ERα. The contribution of this


process to the anti-diabetic properties of E2 is substantial, as endothelial ERα deletion fully negated the antidiabetic actions of the hormone in HFD-fed ovariectomized female mice.


Springboarding from the findings of non-biased interrogation of the ERα interactome and translatome in the endothelium, it was further revealed in cell culture that ERα governance of


endothelial insulin transport requires coupled nuclear and non-nuclear actions on sorting nexin 5 (SNX5). SNX5 is a member of the SNX protein family of cytoplasmic and membrane-associated


proteins that mediate vesicular trafficking of plasma membrane proteins57,58,59. We show that in order for the E2-ERα tandem to enhance endothelial insulin transport, ERα must serve as a


transcription factor upregulating SNX5 expression and also as a means of PI3 kinase activation, which targets SNX5 to join ERα on the plasma membrane. We have determined that SNX5 knockdown


attenuates both E2-stimulated insulin uptake and transcytosis. Since the process of transcytosis requires insulin uptake, these combined findings likely indicate that the SNX5-ERα complex


regulates insulin endocytosis. Similar to the SNX5-related mechanisms revealed in cell culture, there are now two sets of observations that begin to address the participation of both nuclear


and non-nuclear processes involving ERα promotion of insulin sensitivity in vivo. In prior work, we demonstrated that non-nuclear ER activation alone with EDC in ovariectomized, HFD-fed


female mice does not afford the protection from insulin resistance observed with E2 treatment52. In the present work, we demonstrate that the translation of the SNX5 gene is increased by E2


in the skeletal muscle microvascular endothelium in vivo (Fig. 5f). Thus, the nuclear ERα action found to be critical to endothelial insulin transport in culture is relevant in vivo, and


non-nuclear ERα action is insufficient in vivo. Along with deepening our understanding of the metabolic actions of estrogens, the present work identifies a physiologic process that promotes


endothelial insulin transport to skeletal muscle to foster normal glucose homeostasis. The present discoveries further raise the possibility that therapeutic strategies can be developed


which will combat type 2 diabetes by enhancing endothelial cell insulin transport, thereby increasing insulin delivery and ultimately glucose disposal in skeletal muscle. METHODS MOUSE


MODELS Mice with endothelial cell-specific deletion of ERα (ERαΔEC) were generated by crossing ERα floxed mice (ERαfl/fl)66 with vascular endothelial cadherin promoter-driven Cre mice


(VECad-Cre)29,67. Cre-negative ERαfl/fl mice served as controls. To assess the efficiency and cell-specificity of ERα deletion from endothelial cells in vivo, primary endothelial cells were


cultured from mouse aorta as previously described68, and then further purified using rat anti-mouse CD31 (1:500, ThermoFisher cat. No. RM5200) and covalently bound sheep anti-rat IgG on


magnetic beads (Dynabeads, ThermoFisher). Myeloid lineage cells were purified from bone marrow using biotinylated rat anti-mouse Mac-1 antibody (1:200, BD Biosciences cat. No. 557395),


streptavidin particles (BD Biosciences), and Easysep kits (STEMCELL Technologies)68. RNA was isolated from the endothelial cells or myeloid lineage cells, and quantitative RT-PCR for mouse


ERα was performed, expressing steady-state ERα transcript levels relative to mouse hypoxanthine guanine phosphoribosyl transferase (HPRT) endogenous control52, and then normalized to


abundance in Cre negative ERαfl/fl mice. Quantitative RT-PCR TaqMan assay information is provided in Supplementary Table 4. Experiments were performed in both male and female mice. The mice


were housed at 23 °C with light cycles of 12 h of light beginning at 6:00am and 12 h of dark beginning at 6:00 pm, humidity was 30-70%, and food and H2O were provided ad libitum. Beginning


at 5 weeks of age, the males received a standard chow (Harlan Teklad 2016, 12% calories from fat) and the females received a high-fat diet (HFD; Harlan Teklad TD88137, 42% calories from fat,


0.2% cholesterol) for 12 weeks. The females also underwent ovariectomy at 6 weeks of age, at which time treatment was initiated with vehicle or E2 at 6 ug/d using subcutaneously implanted


pellets (Innovative Research of America). Effective loss of endogenous estrogen action versus successful E2 delivery was confirmed at the time of euthanasia by measurement of uterine wet


weights. Additional experiments were performed in female mice to evaluate the effects of E2 on actively translated genes in the skeletal muscle microvascular endothelium. This was


accomplished by crossing mice in which the Rosa26 locus has been targeted with a construct containing a floxed stop codon and GFP-_Rpl10a_, which encodes the ribosomal protein L10a with a


GFP tag (Rosa26fsTRAP)69, with VECad-Cre mice to enact endothelial cell-specific expression of GFP-L10a (designated GFP-L10aEC). The female GFP-L10aEC mice were ovariectomized at 6 weeks of


age and treated with vehicle versus E2 pellets as described above for 4 weeks. All animals were treated and cared for in accordance with the Guide for the Care and Use of Laboratory Animals


[National Institutes of Health (NIH), Revised 2011], and the Institutional Animal Care and Use Committee of the University of Texas Southwestern Medical Center approved all experiments.


EVALUATION OF ADIPOSITY Fat mass and lean body mass were determined by NMR (Minispec NMR Analyzer; Bruker). In select experiments, at the time of euthanasia subcutaneous white adipose tissue


(WAT) was quantified by determining the weight of the inguinal fat pad, and visceral WAT was quantified by determining the weight of the gonadal fat pad70. EVALUATION OF GLUCOSE HOMEOSTASIS


Following fasting for 4–6 h, glucose tolerance tests (GTT) and insulin tolerance tests (ITT) were performed in response to an IP injection with D-glucose (1 g/kg BW) or insulin (1unit/kg


BW), respectively71. Tail vein blood samples were obtained at the indicated times for plasma glucose measurement by glucometer (ONE TOUCH Ultra2, Johnson & Johnson). In select studies,


homeostasis model assessment of insulin resistance (HOMA-IR) was determined as previously described to assess relative insulin sensitivity70, or hyperinsulinemic-euglycemic clamps were


performed using our established approach48,71. In brief, in the clamps hyperinsulinemia was initiated with a primed continuous infusion of insulin (20 mU/kg/min), while a variable infusion


of 50% dextrose and monitoring of blood glucose every 5 min by glucometer allowed for the achievement of a targeted blood glucose level between 115-125 mg/dl. The glucose infusion rate was


calculated. To evaluate pancreatic insulin secretion, mice fasted for 16 h were administered an IP injection of D-glucose (3 g/kg BW), and plasma samples were obtained at baseline and 2, 5,


15, and 30 min post-injection. Plasma insulin concentrations were determined by ELISA (Crystal Chem Inc. #90080 Ver. 15)71. To evaluate relative hepatic insulin sensitivity by assessing


hepatic gluconeogenesis, pyruvate tolerance tests (PTT) were performed. Mice were fasted for 4 h, and changes in plasma glucose were evaluated over 120 min following an IP injection of


pyruvate (2 g/kg body weight)70. QUANTITATIVE REAL-TIME PCR FOR ADIPOSE GENES RNA from inguinal (subcutaneous WAT) and gonadal (visceral WAT) fat pads was isolated using RNAzol RT reagent


(Sigma) according to the manufacturer’s instructions, and cDNA was generated from 2 µg total RNA using the High Capacity RNA-to-cDNA Kit (Applied Biosystems). Flk1, PECAM-1, F4/80, CD11,


IL-6, and TNFα gene expression was evaluated by quantitative RT-PCR as previously described (Supplementary Table 4)72. HPRT expression was assessed in parallel to indicate total RNA


abundance, and relative expression was determined by the 2−∆∆CT method and is reported normalized to the abundance of the transcript in the control group samples. SKELETAL MUSCLE GLUCOSE


UPTAKE, INSULIN SIGNALING, AND INSULIN DELIVERY Skeletal muscle glucose uptake was measured in vivo as previously reported73,74. Briefly, fasted mice were injected IP with


2-deoxy-[3H]glucose ([3H]-2-DOG, Amersham; 2 g/kg; 10 µCi/mouse) mixed with dextrose (20%), and blood glucose was measured at 0–90 min. The glucose-specific activity


(disintegrations/min/µmol) was calculated by dividing the radioactivity by the glucose concentration, and the area under the curve (AUC) was integrated for the duration of the experiment (90


 min). Skeletal muscle (soleus and gastrocnemius) was harvested at 90 min, and the specific accumulation of [3H]-2-DOG was calculated by dividing the radioactive counts (disintegrations/min)


by the integrated glucose-specific activity (AUC) and the sample protein content (Bradford assay, Bio Rad Laboratories). To evaluate skeletal muscle insulin delivery and signaling to Akt,


fasted mice received tail vein injections with vehicle (saline) or bovine insulin (1 unit/kg BW), 5 min later the soleus and gastrocnemius were harvested and homogenized in PBS, and protein


content was quantified (Bradford, BioRad). Phosphorylated Akt and total Akt were detected by immunoblotting using anti-phospho-Akt (S473,1:1000, Cell Signaling cat. No. 9271), anti-Akt


(1:1000, Cell Signaling cat. No. 9272) and anti-GAPDH antibodies (1:2500, Santa Cruz Biotech cat. No. 47724). Insulin abundance was measured by ELISA (Crystal Chem Inc. cat. No. 90095). The


ELISA detects both mouse and bovine insulin, with 2.1-fold greater sensitivity for bovine versus mouse insulin48. To determine if the impact of endothelial ERα deletion on skeletal muscle


glucose uptake entails processes limited to the endothelium, ex vivo studies of insulin-stimulated glucose uptake were performed in isolated soleus and extensor digitorum longus muscle using


our previously established methods71. Briefly, the muscles were excised and incubated at 30 °C in the absence or presence of 2 nmol/L insulin for 40 min in Krebs-Henseleit buffer


supplemented with 5 mmol/L Hepes, 0.1% BSA, and 2 mmol/L pyruvate. Muscles were further incubated for an additional 20 min in Krebs-Henseleit buffer containing 1 mmol/L [3H]2-DOG (2.5 


mCi/mL) and 19 mmol/L [14C]mannitol (0.7 mCi/mL) at 30 °C. After incubation, muscles were digested in 0.5 mol/L NaOH, and extracellular space and intracellular 2-deoxyglucose concentrations


were determined. CONTRAST-ENHANCED ULTRASOUND (CEUS) OF SKELETAL MUSCLE MICROVASCULATURE CEUS was performed on the proximal hind limb adductor muscle group (adductor magnus and


semimembranosus) to determine the microvascular response to a 2-hour hyperinsulinemic-euglycemic clamp entailing an insulin infusion at 20 mU/Kg/min and maintenance of blood glucose at


115–125 mg/dl as previously described46,48. In brief, under isoflurane anesthesia a right jugular vein catheter was placed, and following stabilization the muscle was imaged using a Siemens


Sequoia ultrasound system equipped with a high-frequency 15L8 transducer and a microbubble (MB) contrast agent–sensitive imaging mode (cadence pulse sequencing, CPS) with a transmit


frequency of 10 MHz. CEUS imaging was performed at baseline and at the end of the clamp, immediately before and for 10 min after a 1 min, constant rate-controlled i.v. infusion of a


lipid-shelled perfluorocarbon-based MB contrast agent (Advanced Microbubbles Laboratories). The 1-min constant infusion avoids issues with microbubble floatation in the infusion syringe and


catheter tubing during a longer infusion. Data were recorded and processed offline using prior methods48. An automated region-of-interest (ROI) selection was performed which includes only


the vessels detected by MB signal in the skeletal muscle depicted in the CEUS image after deleting the tissue scatter signal47. The ultrasound transducer was fixed for the entire study


including the repeat in vivo ultrasound imaging sessions. After the selection of the initial ROI from the baseline imaging session, the same area was superimposed and maintained for all


repeat quantifications. With this approach any increases in tissue perfusion are due to microvascular recruitment in response to the insulin clamp as a signal from the larger vessels is the


same for each imaging session/time point. Average time-intensity curves were generated and analyzed to extract parametric measurements of capillary blood volume or CBV (peak intensity,


_Ipk_), microvascular blood flow or MBF (time to peak intensity, _Tpk_; and wash-in rate, _WIR_), and both CBV and MBF (area under the curve, _AUC_; and wash-out rate,


_WOR_)43,44,45,46,47,48. ELECTRON MICROSCOPY OF SKELETAL MUSCLE CAPILLARY ENDOTHELIUM Male mice (_n_ = 6/group) were fed standard chow beginning at weaning, and at 17 weeks of age the


gastrocnemius was harvested for electron microscopic analysis. Tissues were fixed in glutaraldehyde (2.5% in cacodylate buffer pH7.4) followed by osmium tetroxide (1% in cacodylate buffer).


Six capillaries were imaged per mouse using a JEOL 1400 Plus electron micrograph. Vesicles were designated as associated with the luminal plasma membrane or the abluminal plasma membrane, or


localized intracellularly. The number of vesicles was manually counted and divided by the entire area of the endothelium. For the assessment of vesicle area, three randomly chosen regions


were selected per capillary (6 capillaries/mouse, a total of 18 areas/mouse), and the area of each vesicle was measured manually using Image J. ENDOTHELIAL CELL INSULIN TRANSPORT AND GENE


EXPRESSION Human aortic endothelial cells (HAEC) were purchased from Lonza and maintained in EGM-2 and Endothelial Growth Medium with added growth factors (Lonza). Human skeletal muscle


endothelial cells (HSMEC) were obtained from Cell Biologics and were maintained in Complete Human Endothelial Cell Medium with added growth factors (Cell Biologics). Cells were used within 3


to 6 passages. To strengthen the parallel observations in HSMEC and in vivo in mice, select experiments were performed using primary microvascular endothelial cells isolated from the


skeletal muscles (soleus and EDL) of 5-6 week-old male ERαfl/fl and ERαΔEC mice. Briefly, following the dissociation of the tissue with Collagenase IV (0.2% (w/v) and Dispase (1.25 U/ml),


the cell pellet was treated to lyse red blood cells, CD31 MicroBeads (Miltenyi Biotech) were added to resuspended cells, and the CD31+ fraction was isolated using a magnetic separator75.


Prior to the study of insulin transport the cells were placed in media lacking growth factors with 1% FBS added for 18 h. To assess insulin uptake the cells were placed in media containing


vehicle or E2 (10−8 M) and FITC-conjugated insulin (5 × 10−5 M, Sigma-Aldrich) for 30 min at 37 °C and immunofluoresence was detected by microscopy (NIKON Eclipse TE2000, ×20 magnification)


or by POLARstar Omega plate reader (BMG LABTECH). To evaluate the participation of ERα or PI3 kinase, the cells were treated with vehicle or MPP (10−5 M) or LY294002 (5 × 10−5 M) during a


120 min preincubation at 37 °C and during the incubations with FITC-insulin. In select experiments, the requirement for SNX5 in E2-stimulated insulin uptake was interrogated in HAEC and


HSMEC. Cells were transfected with 5 nM control siRNA (Dharmacon, cat. No. D-001810-02-20) or siRNA targeting SX5 (Silencer Select, ThermoFisher cat. No. 439240), and insulin uptake was


studied 24 h later. Effective knockdown of SX5 was confirmed by immunoblotting (1:1000, Abcam cat. No. ab5983). To assess effects of SNX5 on other key participants in endothelial insulin


transport, additional experiments in both HMEC and HSMEC evaluated how SNX5 knockdown impacted IR, IRS-1 and IRS-2 expression using immunoblotting (1:1000, Cell Signaling, cat. No. 234135,


3407T, and 4502, respectively). To exclude off-target effects of SNX5 RNAi, SNX5 expression was reconstituted in HAEC in which siRNA knockdown was previously performed. The siRNA knockdown


proceeded for 24 h and the next day the cells were transfected with concentrated lentiviral particles (Lenti-X Concentrator TAKARA, cat#631232) encoding either eGFP control or SNX5.


E2-stimulated insulin uptake was tested 18 h later. Insulin transcytosis was studied as previously described68. Cells were seeded onto Transwell inserts (6.5 mm diameter, 3 µm pore size,


polycarbonate membrane inserts, Sigma Aldrich) treated with 100ug/ml collagen I (BD Bioscience), and transendothelial electrical resistance (TEER) was monitored daily for 4-6d until studies


were performed using an epithelial volt-ohmmeter (EVOM, World Precision Instruments) to confirm the establishment of a confluent monolayer51. FITC-insulin (5 × 10−8 M, Sigma-Aldrich) was


introduced into the upper chamber along with vehicle or E2(10−8 M), and the cells were incubated for 30 min at 37 °C. At the end of the incubation, the FITC-insulin in the lower chamber was


quantified using a fluorimeter (POLARstar Omega, BMG LABTECH), and the percentage of insulin initially placed in the upper chamber transported to the bottom chamber was calculated.


Incubations with FITC-dextran (M.W. 4000, Sigma Aldrich, 60 min) were then performed, and less than 5% of FITC-dextran added to the upper chamber was detected in the lower chamber indicating


negligible paracellular transport. The stimulation of FITC-insulin transcytosis by E2 was decreased by 84% by the addition of excess unlabeled insulin (5 × 10−4 M), revealing insulin


receptor dependence and confirming that the process entails transcytosis. The participation of ERα or PI3 kinase in insulin transcytosis was evaluated using preincubation and incubation with


vehicle or MPP or LY294002 as described for the insulin uptake studies. In select insulin transcytosis studies the effects of E2 were compared to those of the estrogen dendrimer conjugate


(EDC), which selectively activates non-nuclear estrogen receptors26. Cells were treated with vehicle, E2 (10−8 M), dendrimer control at a concentration equivalent to EDC, or EDC providing


tethered E2 at a concentration equivalent to 10−8 M free E2. In additional experiments insulin transcytosis was evaluated in cells infected with a control lentiviral construct versus a


lentivirus harboring shRNA targeting SNX5. Effective shRNA-based knockdown of SNX5 was confirmed by immunoblotting. To investigate the mechanism by which ERα modulates SNX5 expression, the


receptor was silenced in HAEC by RNAi (Silencer Select, ThermoFisher cat. No. 4392420) confirming effective knockdown by immunoblotting, and in select samples ERα expression was


reconstituted with either wild-type ERα (WT), mutant ERα lacking nuclear localization signals 2 and 3 (ERαΔ250-274) or mutant ERα lacking the DNA binding domain (ERαΔ185-251)60,61. Similar


to what is described above for SNX5 reconstitution, following siRNA knockdown of ERα the cells were transfected with lentiviruses encoding eGFP control, WT ERα, ERαΔ250-274 or ERαΔ185-251,


and 18 h later E2 stimulation of SNX5 expression uptake was tested. Effective ERα knockdown and reconstitution were confirmed by immunoblotting (1:1000, SantaCruz Biotech. cat. No. sc-8002).


PREPARATION OF ESTROGEN DENDRIMER CONJUGATE (EDC) EDC was prepared according to prior methods76,77,78. Briefly, a benzaldehyde derivative of 17α-ethinylestradiol was attached onto the


primary amine groups of a G-6 poly(amido)amine (PAMAM) dendrimer by reductive amination with sodium borohydride in methanol. Small molecule reactants and products were removed by repeated (4


times) ultrafiltration through an Amicon centrifugal filter (Ultra-15, cutoff 30 K) using methanol. The composition of the EDC (average number of estradiol units per PAMAM dendrimer) was


determined by Matrix-assisted laser desorption ionization time of flight (Maldi-TOF) mass spectrometry. For the preparation used in this study, there were an average of 20 estradiol


molecules per molecule of dendrimer, with a polydispersity index of 1.02. The PAMAM dendrimer G6 control (DEN) was simply the G-6 PAMAM without conjugated estradiol molecules.


CO-IMMUNOPRECIPITATION, PLASMA MEMBRANE RECRUITMENT, IMMUNOBLOT ANALYSES Following previously described methods79, in co-immunoprecipitation experiments cells were treated with vehicle


versus E2 (10−8M) for 30 min and then lysed with ice-cold buffer containing 1% Triton X-100, 100 mM NaCl2, 150 mM Tris-HCl, 1 mM CaCl2, 1 mM MgCl2 and protease inhibitors (Roche Diagnostics)


at pH 8.0. Cell lysates were placed on ice for 15 min and then centrifuged at 18,000 × _g_ for 10 min at 4 °C, and supernatants were incubated with protein A beads coated with anti-ERα


antibody (F-10, Santa Cruz Biotech) or matching IgG subclass mock for 3 h at 4 °C with continued rotation. Beads were washed three times and immunoprecipitated complexes were extracted and


analyzed by immunoblot analysis or by mass spectrometry. The immunoblot analyses, which included samples of cell lysates representing the co-immunoprecipitation inputs, were performed for


ERα and SNX5. In studies of plasma membrane targeting, HAEC were treated 5 min with vehicle, E2 (10−8M), or E2 preceded by pretreatment with LY294002 (5 × 10−5 M, 60 min), and plasma


membranes were isolated from total cellular membranes by phase separation (Abcam plasma membrane extraction kit; ab65400). Following treatment, the cells were washed with ice cold PBS and


pelleted twice, and then homogenized with an ice-cold Dounce apparatus. The resulting whole cell lysate was centrifuged at 700 × _g_ for 10 min at 4 °C to pellet nuclei and cell debris, and


the supernatant was centrifuged at 10,000 × _g_ for 30 min at 4 °C to yield a cytosol fraction (supernatant) and total cellular membranes (pellet). The total membrane pellet was subjected to


phase separation twice with centrifugation at 1000 × _g_ for 5 min at 4 °C, the yield of plasma membrane was increased by sample dilution with 5 volumes of water and overnight incubation at


4 °C, and the final plasma membrane pellet was obtained by centrifugation at 21,000 × _g_ for 10 min at 4 °C. The resulting cellular sub-fractions were immunoblotted for GAPDH (marker for


cytosol), calnexin (marker for total membranes; 1:1000, Santa Cruz Biotech. cat. No. sc-23954), VE-cadherin (marker for plasma membrane; 1:1000, Santa Cruz Biotech. cat. No. sc-9989), ERα


and SNX5. LIQUID CHROMATOGRAPHY/TANDEM MASS SPECTROMETRY (LC/MS-MS) To evaluate changes in the ERα interactome in endothelial cells (HAEC) treated with vehicle versus E2 (10−8 M for 30 min),


ERα was immunoprecipitated including mock IgG controls, and the associated proteins were evaluated by liquid chromatography/tandem mass spectrometry (LC/MS-MS)79. Three biological


replicates were used for each condition. Following protein separation by sodium dodecyl sulfate polyacrylamide gel electrophoresis, gel samples were digested overnight with trypsin (Pierce)


followed by reduction and alkylation with dithiothreitol and iodoacetamide (Sigma-Aldrich). After solid-phase extraction cleanup with Oasis HLB plates (Waters), the samples were analyzed by


LC/MS-MS using an Orbitrap Fusion Lumos mass spectrometer (Thermo Electron) coupled to an Ultimate 3000 RSLC-Nano liquid chromatography system (Dionex). Samples were injected onto a 75 um


i.d., 75-cm long EasySpray column (Thermo) and eluted with a gradient from 0 to 28% buffer B over 90 min. Buffer A contained 2% (v/v) ACN and 0.1% formic acid in water, and buffer B


contained 80% (v/v) ACN, 10% (v/v) trifluoroethanol, and 0.1% formic acid in water. The mass spectrometer operated in positive ion mode with a source voltage of 1.5 kV and an ion transfer


tube temperature of 275 °C. MS scans were acquired at 120,000 resolution in the Orbitrap and up to 10 MS/MS spectra were obtained in the ion trap for each full spectrum acquired using


higher-energy collisional dissociation (HCD) for ions with charges 2-7. Dynamic exclusion was set for 25 s after an ion was selected for fragmentation. Raw MS data files were analyzed using


Proteome Discoverer v2.4 SP1 (Thermo), with peptide identification performed using Sequest HT searching against the human-reviewed protein database from UniProt (downloaded April 8, 2022,


20361 entries). Fragment and precursor tolerances of 10 ppm and 0.6 Da were specified, and three missed cleavages were allowed. Carbamidomethylation of Cys was set as a fixed modification,


with oxidation of Met set as a variable modification. The false-discovery rate (FDR) cutoff was 1% for all peptides. The statistical analysis of the mass spectrometry data was performed


using the Differential Enrichment analysis of Proteomics data (DEP)80 analysis workflow package (version 1.12.0), R version 4.0.2. Proteins found in 2 out of 3 replicates per condition were


filtered for further downstream analysis. Variance Stabilizing Normalization was performed to normalize the intensities of the filtered proteins followed by “MinProb” based missing value


imputation. Differential expression testing of conditions was performed using limma empirical Bayes statistics by constructing linear models for proteins. FDR values were higher than


expected, primarily due to variability between samples. Therefore, for significance (_P_ < 0.05) in proteomics results we employed non-adjusted _P_ values. To further identify the


proteins of interest, we only filtered proteins significantly changed with anti-ERα + E2 treatment compared to anti-ERα + vehicle treatment but not significantly changed in the comparison


between Mock IgG + E2 treatment vs Mock IgG + vehicle treatment. IN VIVO ENDOTHELIAL CELL TRAPSEQ Active gene translation was evaluated in the skeletal muscle microvascular endothelial cells


of female GFP-L10aEC mice treated with vehicle versus E2 subcutaneous pellets post-ovariectomy for 4 weeks. Skeletal muscle (gastrocnemius and soleus) was harvested and ribosome-bound RNA


from the muscle endothelium (TRAP RNA) was obtained by anti-GFP immunoprecipitation for 3 h at 4 °C using methods modified from those previously described69. There were 6 biological


replicates per study group. RNAseq was then performed employing the TRAP RNA by the UT Southwestern McDermott Center Sequencing Core. Briefly, using the ARTseq/TruSeq Ribo Profile Library


Preparation Kit (Illumina), RNA-seq libraries were prepared from 500 ng of ribosome-associated RNA. Effective library preparation was validated on a 2100 Bioanalyzer (Agilent Technologies),


and after normalization, 30-40 million reads per library was sequenced on an Illumina NextSeq 500 using 150 nucleotide paired-end chemistry. Image intensities were processed using NextSeq


500 Control Software (Illumina) with default settings. Raw data were de-multiplexed and converted to fastq files using bcl2fastq (v2.17). The fastq files were checked for quality using


fastqc (v0.11.2) (http://www.bioinformatics.babraham.ac.uk/projects/fastqc) and fastq_screen (v0.4.4) (http://www.bioinformatics.babraham.ac.uk/projects/fastq_screen). Sequencing reads were


mapped to mm10 reference genome (from igenomes) using STAR81, counted using featureCounts82 and normalized using trimmed mean of M values (TMM) methods. Differential expression analysis was


performed using edgeR (version 3.12)83. Functional pathway analysis was performed using the ShinyGo application at ShinyGO 0.77 (sdstate.edu)84 with biological process terms from Biological


Processes of Gene Ontology53. For the overlap with proteomics data with TRAPseq data, we first utilized the databases of Homologene (https://www.ncbi.nlm.nih.gov/homologene) and MGI homology


(http://www.informatics.jax.org/homology.shtml) to convert the TRAPSeq mouse data to human homologs. Venn diagrams were generated for the overlapping genes between LC-MS/MS (FDR < 0.01)


and human homologs of the TRAPseq differentially expressed genes (_p_ \(\le\) 0.05 and log2(counts per million) \(\ge\) 0) (http://bioinformatics.psb.ugent.be/webtools/Venn/). QUANITATIVE


RT-PCR FOR TRAPSEQ To evaluate the effective isolation of endothelial cell ribosomes from mouse skeletal muscle in the TRAPseq protocol, Q-RT-PCR was performed using established techniques85


on whole tissue RNA and endothelial TRAP RNA to detect the myocyte-specific genes myogenin (MyoG), -myosin D1 (MyoD1), and alpha-smooth muscle actin (alphaSMA) (α-SMA), and the


endothelium-specific genes roundabout 4 (Robo4), PECAM-1, and VE-Cadherin (Supplementary Table 4). In both tissues and TRAP RNA the transcript abundance was determined relative to the


housekeeping gene hypoxanthine-guanine phosphoribosyltransferase (HPRT). In experiments in cultured endothelial cells similar methods were employed to evaluate relative SNX5 transcript


abundance relative to HPRT mRNA abundance. STATISTICAL ANALYSIS AND REPRODUCIBILITY Following normality testing by Shapiro–Wilk test, for normally distributed datasets comparisons between


two groups were performed by two-sided Student’s _t_ tests, and differences between more than two groups were evaluated by one-way analysis of variance (ANOVA) with Tukey’s post-hoc testing,


or by two-way ANOVA with Sidak’s post-hoc testing. In the instances in which normality testing failed, non-parametric analyses were performed between two non-paired groups using


Mann–Whitney, between two paired groups using Wilcoxon matched-pairs signed rank test, and between more than two groups by Kruskal–Wallis with Dunn’s post-hoc testing. Findings in cell


culture experiments were replicated in two independent experiments. Values shown are mean ± SEM. Significance was accepted at the 0.05 level of probability. REPORTING SUMMARY Further


information on research design is available in the Nature Portfolio Reporting Summary linked to this article. DATA AVAILABILITY The raw LC/MS-MS data in this study has been uploaded to the


MassIVE data repository with accession number MSV000091095. The raw TRAPseq data in this study were deposited in GEO under record GSE179737. There are no restrictions on data availability.


All differential proteomics data and integrated LC/ MS-MS and TRAPSeq analysis results are included in the Tables provided. Source data are provided with this paper. REFERENCES * Hevener, A.


L., Clegg, D. J. & Mauvais-Jarvis, F. Impaired estrogen receptor action in the pathogenesis of the metabolic syndrome. _Mol. Cell Endocrinol._ 418, 306–321 (2015). CAS  PubMed  PubMed


Central  Google Scholar  * Mauvais-Jarvis, F., Clegg, D. J. & Hevener, A. L. The role of estrogens in control of energy balance and glucose homeostasis. _Endocr. Rev._ 34, 309–338


(2013). CAS  PubMed  PubMed Central  Google Scholar  * Wagner, J. D. et al. Insulin sensitivity and cardiovascular risk factors in ovariectomized monkeys with estradiol alone or combined


with nomegestrol acetate. _J. Clin. Endocrinol. Metab._ 83, 896–901 (1998). CAS  PubMed  Google Scholar  * Kumagai, S., Holmang, A. & Bjorntorp, P. The effects of oestrogen and


progesterone on insulin sensitivity in female rats. _Acta Physiol. Scand._ 149, 91–97 (1993). CAS  PubMed  Google Scholar  * Riant, E. et al. Estrogens protect against high-fat diet-induced


insulin resistance and glucose intolerance in mice. _Endocrinology_ 150, 2109–2117 (2009). CAS  PubMed  Google Scholar  * Osborne, C. K. & Schiff, R. Estrogen-receptor biology:


continuing progress and therapeutic implications. _J. Clin. Oncol._ 23, 1616–1622 (2005). CAS  PubMed  Google Scholar  * Musatov, S. et al. Silencing of estrogen receptor alpha in the


ventromedial nucleus of hypothalamus leads to metabolic syndrome. _Proc. Natl Acad. Sci. USA_ 104, 2501–2506 (2007). ADS  CAS  PubMed  PubMed Central  Google Scholar  * Bryzgalova, G. et al.


Mechanisms of antidiabetogenic and body weight-lowering effects of estrogen in high-fat diet-fed mice. _Am. J. Physiol. Endocrinol. Metab._ 295, E904–E912 (2008). CAS  PubMed  PubMed


Central  Google Scholar  * Ropero, A. B., Alonso-Magdalena, P., Quesada, I. & Nadal, A. The role of estrogen receptors in the control of energy and glucose homeostasis. _Steroids_ 73,


874–879 (2008). CAS  PubMed  Google Scholar  * Barros, R., Gabbi, C., Morani, A., Warner, M. & Gustafsson, J. A. Participation of ER {alpha} and ER {beta} in glucose homeostasis in


skeletal muscle and white adipose tissue. _Am. J. Physiol. Endocrinol. Metab._ 297, E124–E133 (2009). CAS  PubMed  Google Scholar  * Davis, K. E. et al. The sexually dimorphic role of


adipose and adipocyte estrogen receptors in modulating adipose tissue expansion, inflammation, and fibrosis. _Mol. Metab._ 2, 227–242 (2013). CAS  PubMed  PubMed Central  Google Scholar  *


Ribas, V. et al. Myeloid-specific estrogen receptor {alpha} deficiency impairs metabolic homeostasis and accelerates atherosclerotic lesion development. _Proc. Natl Acad. Sci. USA_ 108,


16457–16462 (2011). ADS  CAS  PubMed  PubMed Central  Google Scholar  * Heianza, Y. et al. Effect of postmenopausal status and age at menopause on type 2 diabetes and prediabetes in Japanese


individuals: Toranomon Hospital Health Management Center Study 17 (TOPICS 17). _Diabetes Care_ 36, 4007–4014 (2013). CAS  PubMed  PubMed Central  Google Scholar  * Gurka, M. J., Vishnu, A.,


Santen, R. J. & DeBoer, M. D. Progression of metabolic syndrome severity during the menopausal transition. _J. Am. Heart Assoc._ 5, e003609 (2016). PubMed  PubMed Central  Google


Scholar  * Szmuilowicz, E. D., Stuenkel, C. A. & Seely, E. W. Influence of menopause on diabetes and diabetes risk. _Nat. Rev. Endocrinol._ 5, 553–558 (2009). PubMed  Google Scholar  *


Smith, E. P. et al. Estrogen resistance caused by a mutation in the estrogen-receptor gene in a man. _N. Engl. J. Med._ 331, 1056–1061 (1994). CAS  PubMed  Google Scholar  * Maffei, L. et


al. Dysmetabolic syndrome in a man with a novel mutation of the aromatase gene: effects of testosterone, alendronate, and estradiol treatment. _J. Clin. Endocrinol. Metab._ 89, 61–70 (2004).


CAS  PubMed  Google Scholar  * Rochira, V. et al. Oestradiol replacement treatment and glucose homeostasis in two men with congenital aromatase deficiency: evidence for a role of oestradiol


and sex steroids imbalance on insulin sensitivity in men. _Diabet. Med._ 24, 1491–1495 (2007). CAS  PubMed  Google Scholar  * Takeda, K. et al. Progressive development of insulin resistance


phenotype in male mice with complete aromatase (CYP19) deficiency. _J. Endocrinol._ 176, 237–246 (2003). CAS  PubMed  Google Scholar  * Chambliss, K. L. & Shaul, P. W. Estrogen


modulation of endothelial nitric oxide synthase. _Endocr. Rev._ 23, 665–686 (2002). CAS  PubMed  Google Scholar  * Levin, E. R. Integration of the extranuclear and nuclear actions of


estrogen. _Mol. Endocrinol._ 19, 1951–1959 (2005). CAS  PubMed  Google Scholar  * Lantin-Hermoso, R. L. et al. Estrogen acutely stimulates nitric oxide synthase activity in fetal pulmonary


artery endothelium. _Am. J. Physiol._ 273, L119–L126 (1997). CAS  PubMed  Google Scholar  * Chambliss, K. L. et al. Estrogen receptor alpha and endothelial nitric oxide synthase are


organized into a functional signaling module in caveolae. _Circ. Res._ 87, E44–E52 (2000). CAS  PubMed  Google Scholar  * Gu, P. & Xu, A. Interplay between adipose tissue and blood


vessels in obesity and vascular dysfunction. _Rev. Endocr. Metab. Disord._ 14, 49–58 (2013). CAS  PubMed  Google Scholar  * Scalia, R. The microcirculation in adipose tissue inflammation.


_Rev. Endocr. Metab. Disord._ 14, 69–76 (2013). CAS  PubMed  PubMed Central  Google Scholar  * Chambliss, K. L. et al. Non-nuclear estrogen receptor alpha signaling promotes cardiovascular


protection but not uterine or breast cancer growth in mice. _J. Clin. Invest._ 120, 2319–2330 (2010). CAS  PubMed  PubMed Central  Google Scholar  * Lu, Q. et al. ER alpha rapid signaling is


required for estrogen induced proliferation and migration of vascular endothelial cells. _PLoS. ONE_ 11, e0152807 (2016). PubMed  PubMed Central  Google Scholar  * Nilsson, B. O. Modulation


of the inflammatory response by estrogens with focus on the endothelium and its interactions with leukocytes. _Inflamm. Res._ 56, 269–273 (2007). CAS  PubMed  Google Scholar  * Menazza, S.


et al. Non-nuclear estrogen receptor alpha activation in endothelium reduces cardiac ischemia-reperfusion injury in mice. _J. Mol. Cell Cardiol._ 107, 41–51 (2017). CAS  PubMed  PubMed


Central  Google Scholar  * Manrique, C. et al. Endothelial estrogen receptor-alpha does not protect against vascular stiffness induced by western diet in female mice. _Endocrinology_ 157,


1590–1600 (2016). CAS  PubMed  PubMed Central  Google Scholar  * Talavera-Adame, D. & Dafoe, D. C. Endothelium-derived essential signals involved in pancreas organogenesis. _World J.


Exp. Med._ 5, 40–49 (2015). PubMed  PubMed Central  Google Scholar  * Mohamad, M. et al. Ultrastructure of the liver microcirculation influences hepatic and systemic insulin activity and


provides a mechanism for age-related insulin resistance. _Aging Cell_ 15, 706–715 (2016). CAS  PubMed  PubMed Central  Google Scholar  * DeFronzo, R. A., Ferrannini, E., Hendler, R., Felig,


P. & Wahren, J. Regulation of splanchnic and peripheral glucose uptake by insulin and hyperglycemia in man. _Diabetes_ 32, 35–45 (1983). CAS  PubMed  Google Scholar  * Dawson, D. et al.


Vascular recruitment in skeletal muscle during exercise and hyperinsulinemia assessed by contrast ultrasound. _Am. J. Physiol. Endocrinol. Metab._ 282, E714–E720 (2002). CAS  PubMed  Google


Scholar  * Bryant, N. J., Govers, R. & James, D. E. Regulated transport of the glucose transporter GLUT4. _Nat. Rev. Mol. Cell Biol._ 3, 267–277 (2002). CAS  PubMed  Google Scholar  *


Barrett, E. J. & Liu, Z. The endothelial cell: an “early responder” in the development of insulin resistance. _Rev. Endocr. Metab. Disord._ 14, 21–27 (2013). CAS  PubMed  PubMed Central


  Google Scholar  * Barrett, E. J. et al. The vascular actions of insulin control its delivery to muscle and regulate the rate-limiting step in skeletal muscle insulin action. _Diabetologia_


52, 752–764 (2009). CAS  PubMed  PubMed Central  Google Scholar  * Chiu, J. D. et al. Direct administration of insulin into skeletal muscle reveals that the transport of insulin across the


capillary endothelium limits the time course of insulin to activate glucose disposal. _Diabetes_ 57, 828–835 (2008). CAS  PubMed  Google Scholar  * Miles, P. D. et al. Kinetics of insulin


action in vivo. Identification of rate-limiting steps. _Diabetes_ 44, 947–953 (1995). CAS  PubMed  Google Scholar  * Coggins, M. et al. Physiologic hyperinsulinemia enhances human skeletal


muscle perfusion by capillary recruitment. _Diabetes_ 50, 2682–2690 (2001). CAS  PubMed  Google Scholar  * Vincent, M. A. et al. Microvascular recruitment is an early insulin effect that


regulates skeletal muscle glucose uptake in vivo. _Diabetes_ 53, 1418–1423 (2004). CAS  PubMed  Google Scholar  * Wagenmakers, A. J., Strauss, J. A., Shepherd, S. O., Keske, M. A. &


Cocks, M. Increased muscle blood supply and transendothelial nutrient and insulin transport induced by food intake and exercise: effect of obesity and ageing. _J. Physiol._ 594, 2207–2222


(2016). CAS  PubMed  Google Scholar  * Hoyt, K., Umphrey, H., Lockhart, M., Robbin, M. & Forero-Torres, A. Ultrasound imaging of breast tumor perfusion and neovascular morphology.


_Ultrasound Med. Biol._ 41, 2292–2302 (2015). PubMed  PubMed Central  Google Scholar  * Hoyt, K., Sorace, A. & Saini, R. Quantitative mapping of tumor vascularity using volumetric


contrast-enhanced ultrasound. _Invest. Radiol._ 47, 167–174 (2012). PubMed  PubMed Central  Google Scholar  * Saini, R. & Hoyt, K. Recent developments in dynamic contrast-enhanced


ultrasound imaging of tumor angiogenesis. _Imaging Med._ 6, 41–52 (2014). CAS  PubMed  PubMed Central  Google Scholar  * Ghosh, D. et al. Super-resolution ultrasound imaging of skeletal


muscle microvascular dysfunction in an animal model of type 2 diabetes. _J. Ultrasound Med._ 38, 2589–2599 (2019). PubMed  PubMed Central  Google Scholar  * Oezdemir, I. et al. Multiscale


and morphological analysis of microvascular patterns depicted in contrast-enhanced ultrasound images. _J. Med. Imaging (Bellingham)_ 7, 034001 (2020). PubMed  Google Scholar  * Tanigaki, K.


et al. Hyposialylated IgG activates endothelial IgG receptor FcgammaRIIB to promote obesity-induced insulin resistance. _J. Clin. Invest._ 128, 309–322 (2018). PubMed  Google Scholar  *


Williams, I. M. et al. Transendothelial insulin transport is impaired in skeletal muscle capillaries of obese male mice. _Obesity (Silver Spring)_ 28, 303–314 (2020). CAS  PubMed  Google


Scholar  * Wu, Q., Chambliss, K., Umetani, M., Mineo, C. & Shaul, P. W. Non-nuclear estrogen receptor signaling in the endothelium. _J. Biol. Chem._ 286, 14737–14743 (2011). CAS  PubMed


  PubMed Central  Google Scholar  * Vigh, J. P. et al. Transendothelial electrical resistance measurement across the blood-brain barrier: a critical review of methods. _Micromachines


(Basel)_ 12, 685 (2021). PubMed  Google Scholar  * Chambliss, K. L. et al. Nonnuclear estrogen receptor activation improves hepatic steatosis in female mice. _Endocrinology_ 157, 3731–3741


(2016). CAS  PubMed  PubMed Central  Google Scholar  * Gene Ontology, C. The Gene Ontology resource: enriching a GOld mine. _Nucleic Acids Res._ 49, D325–D334 (2021). Google Scholar  *


Acconcia, F. et al. The extra-nuclear interactome of the estrogen receptors: implications for physiological functions. _Mol. Cell Endocrinol._ 538, 111452 (2021). CAS  PubMed  Google Scholar


  * Stellato, C. et al. Identification of cytoplasmic proteins interacting with unliganded estrogen receptor alpha and beta in human breast cancer cells. _Proteomics_ 15, 1801–1807 (2015).


CAS  PubMed  Google Scholar  * Cirillo, F. et al. Molecular mechanisms of selective estrogen receptor modulator activity in human breast cancer cells: identification of novel nuclear


cofactors of antiestrogen-ERalpha complexes by interaction proteomics. _J. Proteome Res._ 12, 421–431 (2013). CAS  PubMed  Google Scholar  * Teasdale, R. D., Loci, D., Houghton, F.,


Karlsson, L. & Gleeson, P. A. A large family of endosome-localized proteins related to sorting nexin 1. _Biochem. J._ 358, 7–16 (2001). CAS  PubMed  PubMed Central  Google Scholar  *


Worby, C. A. & Dixon, J. E. Sorting out the cellular functions of sorting nexins. _Nat. Rev. Mol. Cell Biol._ 3, 919–931 (2002). CAS  PubMed  Google Scholar  * Haft, C. R., de la Luz, S.


M., Barr, V. A., Haft, D. H. & Taylor, S. I. Identification of a family of sorting nexin molecules and characterization of their association with receptors. _Mol. Cell Biol._ 18,


7278–7287 (1998). CAS  PubMed  PubMed Central  Google Scholar  * Kumar, V. et al. Functional domains of the human estrogen receptor. _Cell_ 51, 941–951 (1987). CAS  PubMed  Google Scholar  *


Ylikomi, T., Bocquel, M. T., Berry, M., Gronemeyer, H. & Chambon, P. Cooperation of proto-signals for nuclear accumulation of estrogen and progesterone receptors. _EMBO J._ 11,


3681–3694 (1992). CAS  PubMed  PubMed Central  Google Scholar  * Merino-Trigo, A. et al. Sorting nexin 5 is localized to a subdomain of the early endosomes and is recruited to the plasma


membrane following EGF stimulation. _J. Cell Sci._ 117, 6413–6424 (2004). CAS  PubMed  Google Scholar  * Li, F. et al. Sorting nexin 5 and dopamine d1 receptor regulate the expression of the


insulin receptor in human renal proximal tubule cells. _Endocrinology_ 156, 2211–2221 (2015). CAS  PubMed  PubMed Central  Google Scholar  * Kubota, T. et al. Impaired insulin signaling in


endothelial cells reduces insulin-induced glucose uptake by skeletal muscle. _Cell Metab._ 13, 294–307 (2011). CAS  PubMed  Google Scholar  * Arnal, J. F. et al. Estrogen receptors and


endothelium. _Arterioscler. Thromb. Vasc. Biol._ 30, 1506–1512 (2010). CAS  PubMed  Google Scholar  * Feng, Y., Manka, D., Wagner, K. U. & Khan, S. A. Estrogen receptor-alpha expression


in the mammary epithelium is required for ductal and alveolar morphogenesis in mice. _Proc. Natl Acad. Sci. USA_ 104, 14718–14723 (2007). ADS  CAS  PubMed  PubMed Central  Google Scholar  *


Alva, J. A. et al. VE-Cadherin-Cre-recombinase transgenic mouse: a tool for lineage analysis and gene deletion in endothelial cells. _Dev. Dyn._ 235, 759–767 (2006). CAS  PubMed  Google


Scholar  * Tanigaki, K. et al. Endothelial Fcgamma receptor IIB activation blunts insulin delivery to skeletal muscle to cause insulin resistance in mice. _Diabetes_ 65, 1996–2005 (2016).


CAS  PubMed  PubMed Central  Google Scholar  * Zhou, P. et al. Interrogating translational efficiency and lineage-specific transcriptomes using ribosome affinity purification. _Proc. Natl


Acad. Sci. USA_ 110, 15395–15400 (2013). ADS  CAS  PubMed  PubMed Central  Google Scholar  * Barrera, J. et al. Bazedoxifene and conjugated estrogen prevent diet-induced obesity, hepatic


steatosis, and type 2 diabetes in mice without impacting the reproductive tract. _Am. J. Physiol. Endocrinol. Metab._ 307, E345–E354 (2014). CAS  PubMed  Google Scholar  * Tanigaki, K. et


al. C-reactive protein causes insulin resistance in mice through Fcgamma receptor IIB-mediated inhibition of skeletal muscle glucose delivery. _Diabetes_ 62, 721–731 (2013). CAS  PubMed 


PubMed Central  Google Scholar  * Huang, L. et al. SR-B1 drives endothelial cell LDL transcytosis via DOCK4 to promote atherosclerosis. _Nature_ 569, 565–569 (2019). ADS  CAS  PubMed  PubMed


Central  Google Scholar  * Tanigaki, K. et al. C-reactive protein causes insulin resistance in mice through Fcgamma receptor IIB-mediated inhibition of skeletal muscle glucose delivery.


_Diabetes_ 62, 721–731 (2012). * Zisman, A. et al. Targeted disruption of the glucose transporter 4 selectively in muscle causes insulin resistance and glucose intolerance. _Nat. Med._ 6,


924–928 (2000). CAS  PubMed  Google Scholar  * Conchinha, N. V. et al. Protocols for endothelial cell isolation from mouse tissues: brain, choroid, lung, and muscle. _STAR Protoc._ 2, 100508


(2021). CAS  PubMed  PubMed Central  Google Scholar  * Harrington, W. R. et al. Estrogen dendrimer conjugates that preferentially activate extranuclear, nongenomic versus genomic pathways


of estrogen action. _Mol. Endocrinol._ 20, 491–502 (2006). CAS  PubMed  Google Scholar  * Kim, S. H. & Katzenellenbogen, J. A. Hormone-PAMAM dendrimer conjugates: polymer dynamics and


tether structure affect ligand access to receptors. _Angew. Chem. Int Ed. Engl._ 45, 7243–7248 (2006). CAS  PubMed  Google Scholar  * Kim, S. H. et al. Ligand accessibility and bioactivity


of a hormone-dendrimer conjugate depend on pH and pH history. _J. Am. Chem. Soc._ 137, 10326–10335 (2015). CAS  PubMed  PubMed Central  Google Scholar  * Sacharidou, A. et al.


Antiphospholipid antibodies induce thrombosis by PP2A activation via apoER2-Dab2-SHC1 complex formation in endothelium. _Blood_ 131, 2097–2110 (2018). CAS  PubMed  PubMed Central  Google


Scholar  * Zhang, X. et al. Proteome-wide identification of ubiquitin interactions using UbIA-MS. _Nat. Protoc._ 13, 530–550 (2018). CAS  PubMed  Google Scholar  * Dobin, A. et al. STAR:


ultrafast universal RNA-seq aligner. _Bioinformatics_ 29, 15–21 (2013). CAS  PubMed  Google Scholar  * Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose


program for assigning sequence reads to genomic features. _Bioinformatics_ 30, 923–930 (2014). CAS  PubMed  Google Scholar  * Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a


Bioconductor package for differential expression analysis of digital gene expression data. _Bioinformatics_ 26, 139–140 (2010). CAS  PubMed  Google Scholar  * Ge, S. X., Jung, D. & Yao,


R. ShinyGO: a graphical gene-set enrichment tool for animals and plants. _Bioinformatics_ 36, 2628–2629 (2020). CAS  PubMed  Google Scholar  * Umetani, M. et al. The cholesterol metabolite


27-hydroxycholesterol promotes atherosclerosis via proinflammatory processes mediated by estrogen receptor alpha. _Cell Metab._ 20, 172–182 (2014). CAS  PubMed  PubMed Central  Google


Scholar  Download references ACKNOWLEDGEMENTS This work was supported by National Institutes of Health grants HL144572 (P.W.S.), DK110127 (C.M.), HL098040 (A.S, P.W.S.), S10OD021685-01A1


(K.L.-P.), DK015556 (J.A.K.), CA220284 (B.S.K., J.A.K.), EB025841 and DK126833 (K.H.), by Breast Cancer Research Foundation grants BCRF20-083 (B.S.K.) and BCRF20-084 (J.A.K.), by the


American Heart Association (19POST34390001, J.P.), and by the Associates First Capital Corporation Distinguished Chair in Pediatrics (P.W.S.). AUTHOR INFORMATION AUTHORS AND AFFILIATIONS *


Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA Anastasia Sacharidou, Ken Chambliss, Jun Peng, 


Jose Barrera, Keiji Tanigaki, Chieko Mineo & Philip W. Shaul * Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA Katherine Luby-Phelps 


& Chieko Mineo * Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080, USA İpek Özdemir, Shashank R. Sirsi & Kenneth Hoyt * University of Cincinnati


Cancer Institute, Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45219, USA Sohaib Khan * Department of Chemistry, University of


Illinois at Urbana-Champaign, Urbana, IL, 61801, USA Sung Hoon Kim & John A. Katzenellenbogen * Departments of Physiology and Cell Biology, University of Illinois at Urbana-Champaign,


Urbana, IL, 61801, USA Benita S. Katzenellenbogen * Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA Mohammed


Kanchwala, Adwait A. Sathe & Chao Xing * Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA Andrew Lemoff * Lyda Hill Department of


Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA Chao Xing Authors * Anastasia Sacharidou View author publications You can also search for this author


inPubMed Google Scholar * Ken Chambliss View author publications You can also search for this author inPubMed Google Scholar * Jun Peng View author publications You can also search for this


author inPubMed Google Scholar * Jose Barrera View author publications You can also search for this author inPubMed Google Scholar * Keiji Tanigaki View author publications You can also


search for this author inPubMed Google Scholar * Katherine Luby-Phelps View author publications You can also search for this author inPubMed Google Scholar * İpek Özdemir View author


publications You can also search for this author inPubMed Google Scholar * Sohaib Khan View author publications You can also search for this author inPubMed Google Scholar * Shashank R.


Sirsi View author publications You can also search for this author inPubMed Google Scholar * Sung Hoon Kim View author publications You can also search for this author inPubMed Google


Scholar * Benita S. Katzenellenbogen View author publications You can also search for this author inPubMed Google Scholar * John A. Katzenellenbogen View author publications You can also


search for this author inPubMed Google Scholar * Mohammed Kanchwala View author publications You can also search for this author inPubMed Google Scholar * Adwait A. Sathe View author


publications You can also search for this author inPubMed Google Scholar * Andrew Lemoff View author publications You can also search for this author inPubMed Google Scholar * Chao Xing View


author publications You can also search for this author inPubMed Google Scholar * Kenneth Hoyt View author publications You can also search for this author inPubMed Google Scholar * Chieko


Mineo View author publications You can also search for this author inPubMed Google Scholar * Philip W. Shaul View author publications You can also search for this author inPubMed Google


Scholar CONTRIBUTIONS In vivo studies of adiposity and glucose homeostasis and related parameters, and tissue analyses were performed by K.C., J.B., and K.T.; contrast-enhanced ultrasound


and its analyses by J.P., I.O., S.S., and K.H.; TRAPseq by A.S.; provision and generation of mouse lines by A.S, K.C., and S.K.; electron microscopy and advice on image analysis by K.L.-P.


and the UT Southwestern Electron Microscopy Core; endothelial cell insulin uptake and transcytosis by A.S.; provision of EDC and advice on its use by S.H.K., B.S.K., and J.A.K.;


coimmunoprecipitation and immunoblotting by A.S.; coimmunoprecipitation and LC/MS-MS by A.S., A.L. and the Protein Core at UT Southwestern; RNAseq by A.S.; statistical analyses by M.K.,


A.A.S., and C.X.; A.S., K.H., C.M., and P.W.S. designed the study; and A.S., K.C., C.M., and P.W.S. prepared and wrote the manuscript. CORRESPONDING AUTHORS Correspondence to Chieko Mineo or


Philip W. Shaul. ETHICS DECLARATIONS COMPETING INTERESTS The authors declare no competing interests. PEER REVIEW PEER REVIEW INFORMATION _Nature Communications_ thanks the anonymous


reviewer(s) for their contribution to the peer review of this work. ADDITIONAL INFORMATION PUBLISHER’S NOTE Springer Nature remains neutral with regard to jurisdictional claims in published


maps and institutional affiliations. SUPPLEMENTARY INFORMATION SUPPLEMENTARY INFORMATION DESCRIPTION OF ADDITIONAL SUPPLEMENTARY FILES SUPPLEMENTARY MOVIE 1 SUPPLEMENTARY MOVIE 2 REPORTING


SUMMARY SOURCE DATA SOURCE DATA RIGHTS AND PERMISSIONS OPEN ACCESS This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing,


adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons


license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a


credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted


use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/. Reprints and permissions ABOUT


THIS ARTICLE CITE THIS ARTICLE Sacharidou, A., Chambliss, K., Peng, J. _et al._ Endothelial ERα promotes glucose tolerance by enhancing endothelial insulin transport to skeletal muscle. _Nat


Commun_ 14, 4989 (2023). https://doi.org/10.1038/s41467-023-40562-w Download citation * Received: 12 August 2021 * Accepted: 01 August 2023 * Published: 17 August 2023 * DOI:


https://doi.org/10.1038/s41467-023-40562-w SHARE THIS ARTICLE Anyone you share the following link with will be able to read this content: Get shareable link Sorry, a shareable link is not


currently available for this article. Copy to clipboard Provided by the Springer Nature SharedIt content-sharing initiative