Abstract
Free full text
Role of PVAT in obesity-related cardiovascular disease through the buffering activity of ATF3
Summary
Thoracic aortic perivascular adipose tissue (PVAT) is an adipose organ exhibiting similarities to brown adipose tissue (BAT), including cellular morphology and thermogenic gene expression. However, whether the PVAT phenotype is indistinguishable from the BAT phenotype in physiological vasculature remains unclear. We demonstrated that PVAT is distinguishable from classical BAT, given its specific vessel-tone-controlling function. Activating transcription factor 3 (ATF3) is a key factor in hypertension. Compared with wild-type mice, ATF3-deficient (ATF3−/−) mice fed a high-fat diet exhibited elevated mean arterial pressure, increased monocyte chemoattractant protein-1 expression and hypertrophy, plus abnormal fatty tissue accumulation in the thoracic aortic PVAT, and enhanced vascular wall tension and vasoconstrictive responses of potassium chloride, U46619, and norepinephrine in isolated aortic rings, which were restored after administration of adeno-associated ATF3 vector. We suggest that PVAT, not BAT, modulates obesity-related vascular dysfunction. ATF3 within PVAT could provide new insights into the pathophysiology of obesity-related cardiovascular diseases.
Introduction
Adipose tissue is commonly categorized into three types with distinct functions, phenotypes, and anatomical localizations.1,2,3 White adipose tissue (WAT) is primary energy storage; brown adipose tissue (BAT) is responsible for energy dissipation during cold exposure; beige or brite (brown-in-white) adipose tissue, found interspersed in WAT, is characterized by a brown-like phenotype.4 These three types of adipose tissues also have endocrine functions and play important roles in systemic metabolic processes, especially in obesity and its comorbidities, such as cardiovascular diseases.4,5,6 The role of adipocytes, especially perivascular adipose tissue (PVAT), was first demonstrated in 1991.7 Their well-established non-uniform morphological structure surrounds blood vessels to support the cardiovascular system.6,7 The morphology of PVAT is not always BAT in mice and humans,1,4,8 and its features depend on the regional location and heterogeneity of the vascular bed.1,4 Unlike white fat, thoracic aortic PVAT has similar mitochondrial and metabolic-related gene expression and exerts thermogenic activity during cold adaptation, thereby regulating vascular tone, relaxation, or constriction.7,9 As for the relationship between PVAT and metabolic disease and vasoconstriction, the anti-constriction ability of PVAT is reportedly attenuated when cardiovascular diseases, such as obesity, diabetes, or early atherosclerosis, are dysfunctional.5,10,11 PVAT is also an active endocrine organ that not only produces adipokine-derived relaxing factor (ADRF) but also induces the secretion of adipocyte-derived contracting factor (ADCF) to maintain homeostasis and contribute to the microenvironment of PVAT.6,11 When the thoracic aortic PVAT releases ADRF or ADCF, ADRF functions like nitric oxide (NO), angiotensin 1–7 (Ang1–7), and adiponectin to dilate blood vessels and protect the vascular walls in hyperlipidemia, local hypoxia, and lipid deposition.12 Although ADCFs function similarly, such as AngII, prostaglandins, and norepinephrine (NE), they mediate vasoconstriction and maintain smooth muscle cell elasticity and efficiency.13
The activating transcription factor (ATF)-3 gene belongs to the ATF/cyclic adenosine monophosphate response element-binding (CREB) group and is a stress-response transcription factor.14,15 ATF3 regulates the function of pancreatic β-cells and reduces the occurrence of diabetes,14,15 promotes lipolysis, and attenuates metabolic dyshomeostasis caused by a high-fat diet (HFD).16 Previous studies have clarified that the pathogenic mechanism of diabetes is related to the abnormal expression of ATF3.15,17 High-fat-induced diabetes and obesity models in mice with ATF3 deletion or deficiency have exhibited significantly reduced pancreatic β-cell function, decreased insulin secretion, and exacerbated metabolic dysfunction.15,16 When the loss of ATF3 was restored by the adenovirus atf3 vector, glucagon expression in αTC-1.6 cells was significantly increased through a negative feedback regulation mechanism to improve blood glucose stability.15,18 When ATF3 is activated, it can enter the nucleus and bind to DNA to inhibit CCAAT/enhancer-binding protein α expression and adipocyte differentiation.16 ATF3 overexpression inhibits not only the expression of adipogenesis-related genes but also carbohydrate-responsive element-binding protein (ChREBP) and lipid biosynthesis enzymes such as stearoyl-CoA desaturase (Scd-1), which promote lipolysis and fat browning.16 Based on gene polymorphisms19 and human gene expression databases to analyze clinically moderate to morbidly obese patients, ATF3 expression in the liver, adipose tissue, and muscle tissue has been found to be significantly lower than that in healthy subjects.16 It has been proven that ATF3 is related to obesity in humans.16,19,20
Although the biological function of ATF3 increases the resistance to obesity by regulating the metabolism of adipocytes and improving the metabolic abnormalities caused by an HFD,16,17 the specific role of ATF3 in obesity-induced hypertension and vascular tone regulation remains to be thoroughly investigated. A critical role of the PVAT structure is that it not only modulates vascular homeostasis but also protects against the variation in blood pressure caused by cardiovascular dysfunction.6,21 Previous reports have demonstrated that an increase in ATF3 activity significantly accelerates adipocyte tissue browning and lipolysis in HFD-induced obese mice.16,17 Although accumulating evidence has led to our understanding of the role of PVAT in modulating vascular tone, its impact on vascular disease, especially on how ATF3 affects blood pressure control in obesity-mediated complications, remains to be clarified. To the best of our knowledge, this study is the first to demonstrate a protective function of ATF3 expressed in the thoracic aorta and to explore whether ATF3 in PVAT regulates the release of ADRF and ADCF to relax vascular smooth muscle cells (VSMCs).
Results
Specific heterogeneity of PVAT is essential for regulating vascular function
To reveal the importance of adipose tissue in different regions and phenotypes in vascular protection in response to pathophysiological conditions and vascular homeostasis, we first performed adipose tissue extraction experiments combined with blood vessel myography in wild-type (WT) obese mice. In strategy 1, inguinal WAT (iWAT) extract was administered at resting tone. Slight vasodilation was noted but with no significance compared with the control solvent (Figures 1A and 1D). Similar results were found for the suprascapular BAT extract, although the extract from suprascapular BAT did not cause vasodilation (Figures 1B and 1E). In strategy 2, inguinal WAT and suprascapular BAT extracts remained unchanged during PhE-induced vasoconstriction (Figures 1A, 1B, 1G, and 1H). In particular, PVAT extracts were given 1, 2.5, 5, and 10 μg/mL of adipose tissue concentration, which efficiently relaxed thoracic aortic rings from WT mice in a concentration-dependent manner (Figures S2). Thoracic aortic PVAT extract significantly induced resting tone vasodilation (Figures 1C and 1F) and further attenuated PhE-induced vasoconstriction in WT control mouse aortae (Figures 1C and 1I). We performed time course experiments to clarify further that thoracic aortic PVAT, but not inguinal WAT and suprascapular BAT, extracts exert vasodilatory effects. The results from Figure 1J demonstrated that thoracic aortic PVAT extract increased vasodilation over time, and the results are consistent with those shown in Figure 1F. Consequently, we draw a preliminary conclusion that only substances extracted from thoracic aortic PVAT, but not from inguinal WAT or suprascapular BAT, could modulate vasoconstriction or dilation.
Hypertensive mice and patients both have reduced ATF3 levels in different organs
To explore the candidate genes that may mediate PVAT-induced vasodilation, we screened the human genomic database from the GEO. In the hypertensive disease model, the expression of ATF3 was significantly altered (Figures 2A and 2B). Compared with the control (normotension) group, the expression of ATF3 in the aorta and kidney segments (Figures 2A and 2B, respectively) was significantly lower in the hypertensive group, whereas ATF3 expression in the heart (Figure 2C) and liver tissues (Figure 2D) was not significantly different. We further evaluated the expression of ATF3 in human peripheral blood cells from healthy and hypertensive subjects (Figure 2E) and found that hypertensive patients had a marked decrease in ATF3 expression compared with healthy volunteer controls (Figure 2E). Based on the GEO results in the aorta and kidney, which plays a central role in regulating arterial blood pressure, ATF3 expression was associated with hypertension in both murine strains and human subjects.
Abnormal blood pressure regulation and weight gain are associated with ATF3 deletion
To further examine whether ATF3 plays an essential role in regulating blood pressure in mice fed an ND or HFD, blood pressure was determined using a non-invasive tail-cuff system. Physiological parameters, pathological examination, and related traits in ATF3-null (ATF3−/−) mice were determined for comparison. ATF3−/− mice fed an HFD, but not a regular diet, showed a higher body area with moderate to severe obesity (Table 1). Food intake was not different between the diet-challenged groups (Table 1); however, the body weight changes were significantly increased in ATF3−/− mice compared with their WT littermates (Figures 3A and 3B). In addition, the fasting glucose levels and hepatic steatosis significantly increased in ATF3−/− mice when the HFD was administered for a long period of 22 weeks (Table 1). Surprisingly, both the systolic (Figure 3C) and diastolic (Figure 3E) blood pressures increased in ND-fed mice but were significantly exacerbated in HFD-fed mice (Figures 3D and 3F). The mean arterial pressure was significantly elevated in ATF3−/− mice and was associated with weight gain (Figures 3G and 3B). Heart rates between the control WT and ATF3−/− mice were not statistically different following the different diets (Figure 3H). The cohort, GEO analysis, and murine model results suggest that ATF3 is a pivotal regulator in hypertension disease.
Table 1
ND | HFD | |||
---|---|---|---|---|
Groups | WT | ATF3−/− | WT | ATF3−/− |
Body weight (g) | 28.9 ± 0.6 | 28.4 ± 0.7 | 48.8 ± 0.7 | 52.4 ± 0.8a |
Body width (cm) | 3.0 ± 0.0 | 3.4 ± 0.1 | 4.1 ± 0.1 | 4.8 ± 0.1 |
Body length (cm) | 9.0 ± 0.4 | 9.2 ± 0.1 | 11.2 ± 0.4 | 9.3 ± 0.8 |
Body area (mm) | 2640.8 ± 12.9 | 2669.9 ± 9.9 | 3193.5 ± 15.9 | 3662.4 ± 19.6a |
Food intake (g/wks) | 21.4 ± 2.3 | 21.1 ± 2.1 | 28.6 ± 2.1 | 25.8 ± 2.4 |
Fasted glucose (mg/dL) | 98.1 ± 4.6 | 131.0 ± 4.5a | 127.3 ± 5.3 | 187.0 ± 10.2a |
Hepatic steatosis | No observations | No observations | Yes (mild) | Yes (moderate) |
Data are mean ± SEM.
Body weight, width, length, area, food intake, and fasting glucose in wild-type (WT) and ATF3−/− mice fed a normal diet (ND) or high-fat diet (HFD) for 22 weeks. The hepatic steatosis score was estimated based on the histological examination. Data are mean ± SEM and p < 0.01 compared with the WT group fed ND or HFD in each experiment (n = 5 per group).
ATF3 deficiency alters the morphology and structure of PVAT in high-fat diet-induced obesity
Recent evidence has demonstrated that obesity causes inflammation and oxidative stress to promote increased PVAT mass and lipid droplet hypertrophy surrounding the aortic artery.11 WT mice fed an HFD showed a decreased ATF3 expression (Figure S3). We found that ATF3−/− mice fed a long-term HFD, but not an ND, showed increased PVAT mass and density expansion, significant lipid deposition, and fat distribution in the surrounding PVAT compared with their WT littermates (Figure 4A). HFD-fed ATF3−/− mice showed a markedly increased PVAT area (Figure 4B), width (Figure 4C), and all PVAT tissue weights (Figure 4D) compared with WT mice fed a regular diet and HFD, respectively (Figures 4A–4D). A histological examination further determined the morphological changes in the aortic specimens. The inner diameter (Figures 4E and 4F) and media area (Figures 4E and 4G) showed no differences between the WT and ATF3−/− mice fed the ND or HFD for 22 weeks (Figures 4E–4G). The adventitia area (Figures 4E and 4H) and A/M ratio were significantly increased in HFD-fed ATF3−/− mice (Figure 4I). The high A/M ratio indicated noticeable early fibrotic changes in the adventitia of vessels and PVAT in ATF3−/− mice (Figure 4I). In contrast, fibrosis-like pathological changes were not observed in the WT littermates (Figure 4I). Despite excessive perivascular adipose accumulation and increased fat mass, the blood vessel diameter and media remained unchanged compared with the WT littermates fed the HFD (Figures 4E–4G). Next, we calculated the PVAT adipocyte cell size in each group (Figures 4E, 4J, and 4K) to demonstrate that ATF3 deletion in mice aggravated HFD-induced (Figure 4K), but not ND-induced, adipocyte hypertrophy (Figure 4J). The analysis showed enlarged lipid sizes and estimated fat to the hypertrophy stage (Figures 4E, 4J, and 4K). HFD-fed ATF3−/− mice showed a marked phenotypic conversion of average to enlarged PVAT adipocyte cell size (Figure 4K). This implied that vascular diameters or media formation were unaltered in HFD-fed ATF3−/−mice (Figures 4E–4G); however, the pathological changes in PVAT adipocyte phenotype may have contributed to the susceptibility of arteries to obesity (Figures 4E, 4H, and 4I). These data confirm that the presence or absence of ATF3 ablation in mice modulates PVAT genetic programming, in turn affecting WAT/BAT balance in HFD-fed mice, leading to pathological changes.
Global ATF3 deficiency is responsive and more sensitive to low concentrations of stimulated vasoconstrictors
To determine whether the deletion of ATF3 alters vasoconstricting agent-stimulated contractility, which is enhanced in the aortic arteries of obese mice, we first investigated the vascular reactivity of VSMCs in healthy WT and global ATF3−/− mice fed an ND and a 60% kcal from fat to test the contractile response. Both PVAT and endothelium of the aortic rings were completely removed before myography. In both groups fed the ND, the contractile responses in isolated VSMCs from ATF3−/− mice to a cumulative KCl concentration did not significantly differ compared with vessels from WT mouse VSMCs (Figures 5A and 5B). KCl-induced vasoconstriction, regardless of the concentration-induced tracing curve or tension induced by the maximal concentration of 120 mM, did not differ between the WT and ATF3−/− mice (Figures 5B and 5C). In contrast, low concentrations of KCl in age-matched ATF3−/− mice fed an HFD led to significantly increased vasoconstriction (Figures 5D and 5E). However, high concentrations of KCl did not substantially affect the tension of KCl-induced maximal contraction (Figure 5F), which was consistent with the results shown in Figure 5C. In parallel, we further investigated the concentration-response curve to the thromboxane A2 analog U46619 for isolated aortic arteries with VSMCs (Figures 5G–5L). There was no difference in U46619-induced concentration-dependent vasoconstriction between the WT and ATF3−/− mice fed an ND (Figures 5G and 5H). In contrast, in ATF3−/− mice fed an HFD, a low concentration of U46619 led to significantly increased vasoconstriction (Figures 5J and 5K). Similar results were found in absolute tension at 1 μM; the maximal concentration induced by U46619 showed no differences in either WT or ATF3−/− mice fed an ND or HFD (Figures 5I and 5L). Furthermore, we tested PhE, an α1-adrenergic receptor agonist, to induce VSMC contraction in WT and ATF3−/−mouse arteries. There was no significant difference in the PhE-induced vascular tone (Figure 5M). This implied that the deletion of ATF3 did not affect KCl-, U46619-, or PhE-induced vasoconstriction in mice fed a regular diet. However, these effects were more sensitive and observed in mice fed an HFD, especially in ATF3-deficient conditions followed by low concentration-induced contractions. In addition, the endothelium-independent relaxation of SNP led to no significant differences in the aortic rings from all groups fed the HFD (Figures 5N and 5O). The results showed that the defect of the ATF3 gene in smooth muscle cells did not affect the maximum vascular tone but increased the sensitivity of blood vessels to low-dose vasoconstrictors. Thus, our data suggest that the presence of ATF3 is important for VSMC-regulated vasoconstrictor-induced contractions during the initial phase of obesity.
PVAT from ATF3-deficient mice potentiates and aggravates the constriction ability of aortic rings in obese mice
As shown in Figure 5, we observed a marked increase in vasoconstrictors with KCl- or U46619-induced vascular contractility in ATF3−/− mice fed an HFD compared with the WT controls. However, these effects did not change in the group with normal dietary consumption. We hypothesized that PVAT might regulate agonist-stimulated contractions, enhanced in HFD-induced obesity in mouse aortic rings. In the perivascular adipose-intact aortic rings (+PVAT/VSMC) of control WT littermates fed the HFD, cumulative concentrations of NE significantly caused vasoconstriction. This NE-induced contractility, however, was markedly enhanced in ATF3-deficient aortic rings with PVAT (Figures 6A and 6C). The removal of PVAT increased NE-induced contractions in both groups of aortic rings (Figures 6B and 6D). Furthermore, contractions in response to NE in ATF3−/− VSMCs were significantly higher than that in WT mice at high concentrations (0.03 nM‒1 μM) of NE induction (Figures 6B and 6D). The NE-induced maximal contractions in rings with PVAT were slight in WT mice (Figures 6A and 6E); however, they were markedly increased in ATF3−/− mice presenting with PVAT, which was indicative of the anticontractile effect of PVAT in the WT littermates, but not in the ATF3-deficient mice (Figures 6A and 6E). Our findings of potentiating contractions induced by NE and NE-induced maximal contractions, as represented by tension, were significantly enhanced in ATF3−/− mice fed an HFD (Figures 6A and 6E). Pearson’s equation was used to correct the data and evaluate the ATF3−/− and WT control mice (Figure 6F). We found that increased PVAT weight was associated with a significant increase in NE-induced vasoconstriction in the WT mice (Figure 6F). In ATF3 deficiency, PVAT weight was positively correlated with NE-induced contractions; however, the differences were not statistically significant (Figure 6F).
ATF3-deficient PVAT is less effective in buffering vasoconstrictor serotonin and attenuating the anticontractile ability in obese HFD-fed mice
To test the hypothesis that PVAT influences buffering activity against vascular contraction, we examined the dose-response curve of 5-HT for aortic arteries with PVAT (+PVAT) and without PVAT (−PVAT). As expected, WT and ATF3−/− vessels with PVAT constricted significantly less than those without PVAT; however, 5-HT-induced vasoconstriction was markedly augmented in ATF3−/− mice (Figures 7A and 7B). In addition, WT mice were fed an HFD for 22 weeks, and contraction in response to 5-HT at low concentrations was significantly lower compared with ATF3-deficient mice when the longitudinal removal of PVAT was performed (Figures 7A and 7C). In the VSMCs of ATF3−/− mice, the sensitivity to low concentrations of 5-HT-induced contractions was substantially greater than that in WT mice (Figures 7A and 7C). These findings were consistent with previous observations in the arterial response to U46619 and KCl. However, the loss of ATF3 in VSMCs was substantially sensitive to vasoconstrictors when mice were fed an HFD. We further calculated the area under the curve (AUC) of the concentration-response curve as arbitrary units (AU). As shown in Figure 7D, the measured total area of the 5-HT-induced contractile pattern showed that the whole area level in WT mice was significantly lower than that in ATF3−/− mice in the presence of PVAT (Figures 7D and 7E). In contrast, responses to 5-HT produced by arteries from WT and ATF3−/− mice without PVAT were not significantly different in the contractile patterns and levels of the total area in VSMCs alone (Figures 7F and 7G). To further investigate the buffering activity of PVAT in the vasoconstriction response, we overlapped the 5-HT-induced contraction profiles to analyze the vascular buffering capacity in arteries with and without PVAT. The dotted line area represents the capacity of vascular buffering for vasoconstriction and the Δabc value obtained by subtracting the total area of VSMCs from PVAT (Figures 7H and 7I). In the WT control, the value of Δabc of the buffering capacity to 5-HT-induced contraction was significantly higher when ATF3 was present in PVAT, which was indicative of an anticontractile effect of PVAT in WT mice (Figures 7H and 7I). However, this anticontractile ability was markedly decreased in the absence of ATF3 in mouse PVAT (Figures 7H and 7I). These results indicated that ATF3 might play an epigenetic role in PVAT, resulting in anticontractile effects via VSMC under HFD conditions.
Restoration of ATF3 expression by AAV-8 reverses HFD-induced body weight gain, PVAT structure abnormality, and adipocyte hypertrophy
As a complementary approach to creating knockout mouse studies, we performed phenotype-rescue analyses in which ATF3-deficient mice received an AAV-mediated gene transfer of ATF3 (AAV8-ATF3). Consistent with a previous report, PVAT extracts from ATF3-deficient mice subjected to AAV8-ATF3 injection restored the mRNA expression of ATF3 (data not shown). Obesity was induced by the HFD in the AAV8-ATF3-ATF3−/− and AAV8-GFP-ATF3−/− mice. Expectedly, AAV8-ATF3-ATF3−/− mice fed an HFD for 22 weeks showed a significant reduction in body weight (Figure S5A), marked decrease in PVAT surrounding the thoracic aorta, and improved adipocyte hypertrophy (Figure S5B). Twenty-two weeks after the intravenous injection of AAV8-ATF3 in the tail vein once per week in HFD-fed ATF3−/− mice, the PVAT area (Figure S5C), width (Figure S5D), and tissue weight (Figure S5E) were lower than those in the ATF3−/− mice subjected to AAV8-GFP (Figures S5C–S5E). The histological analysis of the PVAT structure showed that in the original picture (Figure S5F), the diameter (Figure S5G) and media (Figure S5H) remained unchanged, which was consistent with previous results (Figure 4). ATF3−/− mice injected with AAV8-ATF3 in the adventitia (Figure S5I) showed significantly lower A/M ratios (Figure S5J) than AAV8-GFP-ATF3−/− mice (Figures S5I and S5J). In addition, fibrotic changes were observed and were more marked in AAV8-GFP-ATF3−/− mice; however, the AAV8-ATF3 injection reversed the pathological changes in ATF3−/− mice (Figures S5F and S5J). Although the AAV8-GFP group also presented adipocyte hypertrophy and lipid size enlargement, these effects were significantly reversed following the AAV8-ATF3 injection (Figures S5F and S5K). Thus, we can confirm that ATF3 expression in PVAT plays a key role in regulating the morphology or WAT/BAT distribution in the PVAT.
ATF3 restoration by the AAV8-ATF3 transfection strategy provides vascular benefits to obese mice
We further investigated the vascular contractile response in isolated aortic artery rings with and without PVAT when AAV8-GFP and AAV8-ATF3 injections were restored. In AAV8-GFP mice, aortic rings with PVAT showed an augmented response to NE, as demonstrated by evaluating the sensitivity to vasoconstrictors (Figures S6A, S6B, and S6C). The concentration-effect curves (Figures S6A, S6B, and S6C) and maximal concentration of NE-induced absolute tension (Figure S6D) differed significantly when ATF3 expression was restored following the AAV-ATF3 injection. We observed that the contractions in response to NE in the PVAT (Figure S6B) and VSMCs (Figure S6C) were significantly lower, and the higher absolute tension was markedly reversed by AAV8-ATF3 treatment compared with the AAV8-GFP group (Figure S6D). As expected, AAV8-ATF3-treated group mice also exhibited a decreased sensitivity to NE and reversed HFD-induced increased PVAT mass (Figure S6E). In this series of AAV-restoration experiments in the mouse models, the PVAT weights increased in the AAV8-GFP group. They showed a positive correlation with NE-induced vasoconstriction in aortic arteries (Figure S6E), consistent with the results in Figure 6F.
AAV8-mediated expression of ATF3 reverses buffering activity to 5-HT-induced contractile responses in obese mice
We next investigated the AAV8-ATF3-restored anticontractile response and the influence of the buffering activity of PVAT on vasoconstriction responses stimulated by 5-HT induction. In a similar experimental procedure to that shown in Figure 7, we set up the concentration-effect curve for 5-HT and calculated the AUC to estimate the buffering activity of PVAT. The results showed that PVAT in AAV8-ATF3-ATF3−/− mice, but not in AAV-GFP-ATF3−/− mice, attenuated the development of force contraction induced by 5-HT (Figure S7A). In parallel, the percentage of increased contraction with PVAT was lower, and the response curve significantly decreased in AAV8-ATF3-ATF3−/− mice recorded in intact PVAT rings (Figures S7A and S7B). Surprisingly, aortic rings without PVAT obtained from AAV8-ATF3-ATF3−/− mice fed an HFD showed significantly increased smooth muscle contractions (Figures S7A and S7C). We further examined the buffering of PVAT to 5-HT-induced constriction, the AUC curve as the contractile pattern, and measured the total area as the capacity. Twenty-two weeks after the intravenous injections of AAV8-ATF3 in HFD-fed ATF3−/− mice, the +PVAT (Figures S7D and S7E) and −PVAT (Figures S7F and S7G) groups showed significantly different levels of total area compared with AAV8-GFP-ATF3−/− mice (Figures S7E and S7G). In AAV8-ATF3-ATF3−/− mice, the AUC and Δabc as a function of buffering capacity to 5-HT were significantly higher when ATF3 was restored in mice (Figures S7H and S7I). These results suggest that ATF3 plays a crucial role in mediating the anticontractile effect of PVAT and contributes to vascular contractility and the regulation of blood pressure.
ATF3 regulates vascular reactivity via the PVAT-mediated release of ADRF and ADCF
To clarify the molecular mechanisms underlying the regulation of the PVAT anticontractile response and decreased sensitivity of vasoconstrictor-induced contraction in aortic rings by ATF3, we hypothesized that ATF3 might regulate ADCF or ADRF release in HFD-fed mice. Accordingly, adipokines secreted from adipose tissue may have systemic effects and shift to pro-inflammation as adipose tissue expands during the development of obesity.22 Based on our results, we tested whether significant obesity-induced changes in PVAT-derived relaxation and contraction factors balanced affected vessel contractility. We used a protein array analysis and ImageJ quantification to determine the factors involved in PVAT regulation.23 The protein array analysis of adipokines (Figure 8A) revealed higher plasma levels of monocyte chemoattractant protein-1 (MCP-1) (Figure 8B), C-reactive protein (Figure 8C), and interleukin (IL)-6 (Figure 8D) in ATF3−/− mice than in WT mice. The plasma levels of resistin (Figure 8E), PAI-1 (Figure 8F), and adiponectin (Figure 8G) did not differ between groups. Parallel experiments with immunoblotting (Figures 8H, 8I, 8J, and S8) and RT-PCR (Figures 8K and 8L) were performed. Further evidence showed that adipokine ADCF and ADRF secretion, including ADCF MCP-1 protein (Figure 8H) and mRNA (Figure 8K) expression, was higher in the PVAT tissue of ATF3−/− mice than in WT mice (Figures 8H and 8K). We found no changes in ADRF adiponectin protein production (Figure 8I), but there was a significant difference in the mRNA levels in PVAT (Figure 8L). We determined whether the downregulation of adipoR1 in VSMCs was worse than adiponectin-mediated vasorelaxation. Notably, protein levels of adipoR1 in VSMCs were markedly reduced in ATF3−/− mice fed an HFD (Figure 8J). MCP-1 and adiponectin expression in PVAT were examined using double-immunofluorescence staining. Fluorescence intensity in the PVAT surrounding the vessel was quantified and indicated that the ADCF MCP-1 release and overexpression in ATF3−/− aortic rings with PVAT were much stronger than in WT mice (Figures 8M and 8O). We found that ATF3−/− mice fed an HFD showed significantly increased MCP-1 production and release from the PVAT. However, the lack of ATF3 did not affect adiponectin levels in PVAT from both groups fed the HFD (Figures 8M and 8P). AdipoR1 expression was co-stained with the smooth muscle cell marker SM22 in the medial layer of the vessel, indicating that the adipoR1 fluorescence intensity was significantly decreased in ATF3−/− mice (Figures 8N, 8Q, and 8R). Adiponectin expression in both rings with PVAT was not significantly different (Figure 8P); the expressed adipoR1 in VSMCs was markedly reduced in ATF3−/− mice (Figure 8Q). In contrast, the levels of circulating and tissue location with MCP-1 and decreased adipoR1 in VSMCs, which may be related to vascular tone abnormalities in diet-induced obesity, were significantly higher in ATF3−/− mice (Figures 8O and 8Q). We, therefore, propose that the mechanisms of PVAT in the regulation of vascular homeostasis involve the ATF3-mediated regulation of ADCF MCP-1 but not ADRF adiponectin expression and secretion from adipocyte tissue. Our findings prove that ATF3 protects against high vasoconstrictor-induced sensitivity to contractions. Furthermore, ATF3 function in PVAT significantly improved vascular hyperreactivity and the loss of anticontractile ability in HFD-induced vascular dysfunction. Therefore, these effects of ATF3 regulation may be beneficial in reversing obesity-related vascular complications, such as hypertension (Graph abstract).
Discussion
To the best of our knowledge, this study is the first to provide substantial evidence that the functional regulator ATF3 in PVAT, but not BAT or WAT, is essential for physiological homeostasis in blood vessels, contributing to regulating blood pressure. We used traditional whole-body knockout mice and generated recombinant AAV carrying ATF3 to restore the original function of the ATF3 gene. Mice lacking ATF3 gene expression revealed hypertension, vascular tone regulation abnormalities, and potentiated KCl- and U46619-induced contractility in VSMCs.
ATF3 deletion resulted in a lack of anticontractile ability in PVAT, enhanced sensitivity to vasoconstrictors, and increased ADCF MCP-1 release. These results suggest that ATF3 contributes to vascular variations in HFD-induced obesity and related vascular tone regulation abnormalities. This study identified ATF3 as a promising modulator of obesity-induced cardiovascular complications.
ATF3 is an adaptive response transcription factor widely expressed in various organs, such as the aorta, kidney, liver, and heart.16,20 This is in accordance with the NCBI data in Figure 2. However, the specific role of ATF3 in regulating blood pressure remained unclear. Thus, we propose that the global deletion of ATF3 may cause aggregate vasoconstriction and that mice have hypertensive features, even in the absence of an HFD. This is supported by data obtained from ATF3−/− mice in our study, which displayed increased systolic and diastolic pressures with increased mean arterial pressure (Figure 3). Conversely, Chou et al. previously demonstrated that in ATF3-deficient mice with fructose-induced metabolic syndrome, systolic blood pressure was not significantly elevated in either WT or ATF3−/− mice.24 Notably, different model-induced diseases, diet sources, and experimental periods may yield differences in blood pressure measurements.25 Another possibility is that ATF3 induces chronic adaptation to modulate stress-induced vascular dysfunction.14 The model of HFD-induced obesity or age-dependent vascular abnormality for studying ATF3 function depends on the duration of the experiment. We ruled out changes that may have occurred if the feeding duration was shorter than 22 weeks, as both the systolic and diastolic blood pressures showed trends toward an increase in both HFD groups and were age-dependent in the standard diet groups. However, it is clear if the knockout of ATF3 significantly affects systemic blood pressure regulation and the morphological appearance of body fat distribution. In a human study based on data from the NCBI, hypertensive patients showed decreased ATF3 expression (Figure 2).26 This result supports our finding of the correlation between ATF3 expression and hypertension.26 In addition, this is consistent with previous studies that used microarray analyses of other mouse strains to induce hypertensive disease, which indicated that ATF3 expression in the aorta and kidney is involved in the progression of hypertension.27 Our results support the hypothesis that ATF3 deletion exacerbates blood pressure variation and increases contraction responses to contractile stimuli. A previous study demonstrated that IL-18-deficient mice exhibited blood pressure abnormalities.28 However, there were no unusual morphological differences or apparent infiltrating cell accumulation in the tissues.28 The pathological mechanisms affecting blood pressure include an enlarged heart, thickened vascular wall with fibrotic changes, and endothelial dysfunction.28 We revealed that ATF3−/− mice had impaired blood pressure regulation and vascular dysfunction of the aorta, including in VSMCs and PVAT. Changes in the regulation of blood pressure may be related to altered PVAT function and morphological features, but not changes in the heart. Other possible mechanisms of action in the aorta of ATF3−/− mice include impaired EC function (verified by ACh-induced relaxation) and related molecular target AMPK-mediated eNOS enzyme expression. The possible role of the endothelial function remains to be investigated. Our results indicate that both systolic and diastolic blood pressure in ATF3−/− mice were higher than in their WT littermates, even though there was no apparent PVAT abnormality or differences in PVAT anticontractile ability in ND-fed mice for 22 weeks. Therefore, future studies are needed to elucidate the detailed mechanisms of increased blood pressure in ATF3−/− mice. The renin-angiotensin system (RAS) control of blood pressure regulation has been well established.29 A published result from Chang et al. demonstrated that brown adipocyte-selective Bmal1-deficient (BA-Bmal1-KO) mice were mediated by angiotensinogen expression and increased release of AngII from PVAT, which contributed to the regulation of vasoactivity and blood pressure during the resting phase.29 In our study, however, we did not examine the function of RAS in regulating blood pressure, nor did we propose that the level of AngII in ATF3−/− mice when fed normal chow was higher than that in WT mice. We cannot rule out that the RAS system mediates angiotensinogen expression and that AngII levels in the PVAT of ATF3−/− mice may be significantly increased. Therefore, the possibility that ATF3 also regulates the angiotensinogen, AngII, and AT2 receptor pathways, including Bmal1 gene function in PVAT, warrants further investigation.
The metabolic balance of adipose tissue in the lipolysis and lipogenesis pathways involved in regulating blood pressure and vascular homeostasis by ATF3 is poorly understood. Evidence has shown that increased visceral fat due to metabolic disorders is associated with increased cardiovascular diseases, such as hypertension.30,31 Moreover, the accumulation of visceral white adipose fat due to obesity may result in a stressed and dysfunctional state that leads to the release of proinflammatory mediators, including tumor necrosis factor (TNF)-α and IL-6. These results are significant complications in the cardiovascular system.9,10 Based on previous studies, ATF3 has anti-inflammatory and protective effects in various tissues, including the kidneys, heart, and blood vessels.16,17,32,33,34 According to a previous study, ATF3 inhibits obesity-induced inflammation by targeting adipocytes.16 Here, we found that PVAT lipid composition and droplet size showed significant changes in ATF3−/− mice fed an HFD. Morphological analyses revealed no differences in PVAT between WT and ATF3−/− mice fed an ND. The lack of ATF3 expression significantly promoted PVAT morphological abnormalities and increased its adipocyte size and adventitia fibrosis in HFD feeding. Our results are consistent with those of Cheng et al., suggesting that ATF3 may affect the function of adipocytes,16 especially the PVAT structure, to regulate vascular homeostasis. The possible involvement of the protection of vascular dysfunction by controlling PVAT buffering activity supports our hypothesis that may underlie the ATF3-mediated PVAT morphological alternation mechanisms.
One of the findings of this study was the functional effect of ATF3 on the regulation of vascular tone by PVAT in different diet challenges, especially the HFD. We further investigated whether the anticontractile ability of aortic PVAT is lost in ATF3-deleted mice, which is due to the enhanced generation of ADCF MCP-1 release by the PVAT. Our results demonstrated that the protective effect was impaired by the loss of ATF3 in HFD-induced obesity, suggesting that PVAT plays a protective regulator role in aortic contractions. Similar results were found for the PVAT extract switching. WT PVAT extracts induced significant vasodilation. However, the PVAT extract from ATF3−/− mice caused slight vasodilation in WT aortic rings (Figures S4A and S4B). Thus, it was considered that the harmful effects on the ATF3−/− mouse aorta were caused in an additive manner by the release of ADCF and imbalance of ADRF. We previously discussed that the altered PVAT function in ATF3 deficiency appeared to be related to morphological changes. The loss of PVAT function was associated with the increased arterial tone, with an essential role of ATF3 in regulating these processes. This is further supported by our observation that obese ATF3−/− mice were exacerbated with vasoconstrictor agents. We found that WT PVAT significantly attenuated the vascular responsiveness of aortic arteries to contractile agents, including NE and 5-HT. To reach the direct conclusion that ATF3 plays a role in PVAT, a precise mechanism of action of ATF3 in PVAT will be delineated in future studies using a CRISPR knockout experiment model, siRNA knockdown assay in an ex vivo model, and blood-vessel myography assay.
In parallel, ATF3−/− mice without PVAT were more hyperpolarized in the VSMC membrane by activating the potassium channel activator KCl. The mechanism has been described in Figure 5, where potassium channels are primarily involved only in HFD feeding. Based on previous reports, the potassium channels Kv and KATP could contribute to blood pressure elevations in obesity-related hypertension.35,36 In addition, PVAT from BKCa knockout mice showed decreased anticontractile activity and attenuated adiponectin release.37 We cannot rule out that VSMCs and PVAT BKCa channels mediate the process of PVAT-derived relaxation abolished by ATF3 deletion. Therefore, the thromboxane A2 analog U46619 induces vascular constriction, which depends on the resistance to opening the potassium channel of VSMCs.36 Low concentrations of U46619 significantly increased vasoconstriction in HFD ATF3−/− mice. These results were similar to those for KCl, indicating that the difference in the contractile response to KCl and U46619 between VSMCs and PVAT is involved in potassium channel regulation.35,38 The electrophysiological mechanisms underlying ATF3 involved in different types of potassium channel activation require further investigation.
PVAT releases NO, adiponectin, H2S, and methyl palmitate as ADRFs to regulate blood pressure.6,21,39 Studies in animals and humans have also suggested that ADCFs, such as AngII, reactive oxygen species (ROS), and MCP-1 are associated with hypertension.6,21 Currently, direct evidence from animal models to investigate the anticontractile or contractile roles of PVAT in blood pressure regulation is well investigated. Body fat and renal function are further associated with obesity-induced hypertension,3,31,40 and ATF3 might be involved in those organ regulations. The mechanisms of ATF3 deficiency in blood pressure regulation and the maintenance of vascular homeostasis in cells and tissues are also complicated and, thus far, unclear. We used the obese model of a mouse lacking ATF3 expression in the global system and reported that ATF3−/− mice are more likely to have high blood pressure following a long-term ND and HFD feeding. Our present results further demonstrate that the loss of anticontractile ability to NE and buffering activity to vasoconstrictor 5-HT stimulation in ATF3-deficient mice is due to alterations in the structure and morphology of PVAT; moreover, the enhanced early fibrosis parameter A/M ratio to the absence of ATF3 regulation in the VSMCs when HFD diet challenged. Because ATF3 deletion in VSMCs augmented with low-concentration contractions to KCl and U46619, it is also likely due to an increased sensitivity of the medial VSMCs vasoconstrictor application. A comparison of adiponectin expression in the PVAT of 22-week-old WT and ATF3-deficient mice allowed us to conclude that the ADRF adiponectin levels in both groups had no significant difference but markedly declined in adipoR1 expression in ATF3−/− mice on HFD feeding. The lack of change in gene or protein production does not rule out changes in ATF3-mediated adiponectin secretion.
Our results are consistent with those of previous studies reporting that the PVAT-mediated anticontractile effect is lost and impaired in obese models.6,11 The mechanism seemingly involves the reduced release of the relaxing factors or the release of constricting factors that contribute to the loss of the anticontractile effect of PVAT. To further explore the mechanism of the altered function of ATF3 in PVAT, we measured critical adipokines, including adiponectin, NO, MCP-1, and IL-6, in WT and ATF3−/− mouse PVAT extracts. We found that the protein array analysis significantly elevated ADCF MCP-1 (Figure 8B) release and the proinflammatory adipokines C-reactive protein (Figure 8C) and IL-6 (Figure 8D) in ATF3−/− mice. However, unlike MCP-1, C-reactive protein and IL-6 did not induce significant vasoconstriction in our previous findings. We further explored proteomic and KEGG pathway map analysis, and our preliminary data show ATF3-conducted PVAT function and that the PPAR signaling pathway might be involved in blood pressure regulation (unpublished results). We did not find any significant differences in the release and expression of ADRF adiponectin. Interestingly, adipoR1 expression in VSMCs was significantly lower in ATF3−/− mice. The critical molecules in the mechanism of impaired function of ATF3 deficiency may target the adipoR1 receptor but not adiponectin. In this study, we found that MCP-1 and IL-6 release can be enhanced by ATF3 deficiency, which may cause a marked increase in vasoconstriction in ATF3−/− mice, but not in WT aortic rings. Indeed, reduced AMPK activity, leading to an upregulation of inflammatory cytokines IL-6 and TNF-α, is one of the reasons for exploring the possible role of ATF3 in vascular tone regulation.41,42 It must be noted that there is another target of AMPK in addition to adiponectin, and these cannot be ruled out from local hypoxia and ROS generation in HFD-fed states involved in the loss of the anticontractile effect of PVAT.6,33 In this study, PVAT inflammation, oxidative stress, and adipocyte hypertrophy could also lead to a harmful change in AMPK-mediated NOS production or perhaps act as a compensatory mechanism following adipoR1 expression in the PVAT or VSMCs. These pathological changes may cause severe vascular hyper-responsiveness to the inflamed and injured cardiovascular system. The homeostasis needed for maintaining blood pressure at physiological levels during vascular dysfunction associated with obesity requires ATF3 expression.
Adiponectin is a well-established vasodilator secreted by the adipocytes of the PVAT.39,43 Published evidence suggests that the loss of the buffering activity of PVAT is accomplished by decreased adiponectin secretion.42 MCP-1, an ADCF induced by inflammation and local hypoxia, has been demonstrated to contribute to increased vascular hyperreactivity in obese animals and humans.10,22 Attenuated MCP-1 expression and production have been shown to improve PVAT dysfunction, increasing vascular hyperreactivity. Based on a previous report, MCP-1 and IL-6 mRNA expression is modulated by ATF3 transcription,16,32 which plays a role in the negative feedback regulation of inflammation. Our results showed that adiponectin expression in WT and ATF3−/− mice showed no significant changes; however, adipoR1 expression was significantly decreased in HFD-fed ATF3−/− mice. In addition, MCP-1 released from PVAT was greatly enhanced in obese mice but markedly higher in ATF3−/− mice. Augmented MCP-1 and IL-6 overproduction due to ATF3 dysfunction in ATF3−/− mice PVAT supports this notion. This further highlights the importance of ATF3 in maintaining vascular health by controlling ADCF and ADRF release.
Conclusion
From animal links to human studies, our findings suggest that ATF3 could serve as a critical target for regulating hypertensive vascular dysfunction during obesity. In summary, this study demonstrated that obesity induces dramatic PVAT morphological changes in ATF3−/− mice, which are associated with increased sensitivity to vasoconstrictors and attenuated anticontractile activity in the aorta. We also found that in obese ATF3−/− mice, augmented vasoconstriction and increased vascular tone could be aggravated by PVAT-derived ADCF MCP-1 release, suggesting that the release and expression of ADCF, but not ADRF, could contribute to an increased risk of developing hypertension in obese mice. Notably, the imbalance between ADCF and ADRF, especially the ADCF principal function, may be mediated by ATF3 regulation. These functional effects of the vascular regulation of ATF3 could cause blood pressure variations in humans with obesity. ATF3 plays a critical role in maintaining the anticontractile responses and buffering capacity of PVAT, attenuates ADCF MCP-1 release, and protects vascular homeostasis when faced with vasoconstrictor stimulation regulating blood pressure variation in obesity-induced cardiovascular dysfunction. Further research is needed to investigate the detailed mechanisms of ATF3-regulated vascular function and provide preventive strategies for increasing ATF3 activity, with the aim of decreasing obesity-related cardiovascular complications.
Limitations of the study
It remains unclear why ATF3 deficiency promotes hypertension; thus, the detailed mechanisms underlying ATF3 regulation require further examination. The conducting arteries and resistance arteries are quite different; furthermore, in our laboratory setting, the blood-vessel myography system, especially the assessment of vascular functionality in specific knockout mice, is somewhat technically limited because the mouse vasculature can only be analyzed in the aortic region. Isolation of blood vessels from mice in other regions, especially resistant blood vessels, is hindered as the diameter of the myography system is too small and the tension value is too low to interpret. The aortic rings cannot refer to blood pressure change, which is a significant limitation of the study. Hence, we will perform radiotelemetry analysis to determine the systemic blood pressure changes in ATF3−/− and WT mice.
Ethics approval and consent to participate
All animal protocols were approved by the Animal Care and Use Committee of Tzu Chi Hospital in Taiwan (approval no.:108–39 and 110–47).
STARMethods
Key resources table
REAGENT or RESOURCE | SOURCE | IDENTIFIER |
---|---|---|
Antibodies | ||
goat polyclonal anti-MCP-1 antibody | Santa Cruz Biotechnology | Cat# sc-1784; RRID:AB_2070878 |
rabbit monoclonal anti-adiponectin antibody | Invitrogen | Cat# 701148 |
rabbit polyclonal anti-adiponectin receptor-1 (AdipoR1) antibody | GeneTex | Cat# GTX104770; RRID:AB_1240425 |
mouse monoclonal anti-SM22 antibody | Millipore | Cat# MABT167 |
Hilyte Fluor 488-labeled anti-goat IgG for MCP-1 | AnaSpec | Cat# AS-28168-1-H488 |
Hilyte Fluor 488-labeled anti-rabbit IgG for AdipoR1 | AnaSpec | Cat# AS-28176-1-H488 |
Hilyte Fluor 555-labeled anti-rabbit IgG for adiponectin | AnaSpec | Cat# AS-28176-1-H555 |
Hilyte Fluor 555-labeled anti-mouse IgG for SM22 | AnaSpec | Cat# AS-28175-1-H555 |
rabbit polyclonal anti-MCP-1 antibody | Arigo | ARG57649 |
mouse monoclonal anti-actin antibody | Thermo Fisher Scientific | Cat# MA1-24906; RRID:AB_2537000 |
Critical commercial assays | ||
Proteome Profiler Mouse Adipokine Array Kit | R&D Systems | Cat#ARY013 |
ELISA kits for monocyte chemoattractant protein (MCP)-1 | Enzo Life Sciences | Cat#ADI-900-077 |
ELISA kits for mouse adiponectin | MyBioSource | Cat#MBS7725304 |
iScript cDNA Synthesis Kit | Bio-Rad | Cat# 1708890 |
iTaq™ Universal SYBR® Green Supermix | Bio-Rad | Cat# 1725120 |
Deposited data | ||
The microarray experimental data | Gene Expression Omnibus (GEO) of the NCBI | GDS3672; GDS3675; GDS3673; GDS3674; GSE24752 |
Experimental models: Organisms/strains | ||
ATF3−/− mice | Zmuda et al., 201015 | N/A |
C57BL/6 mice | BioLASCO Co., Ltd. Taipei, Taiwan | http://www.biolasco.com.tw/index.php/tw/ |
Oligonucleotides | ||
Mouse adiponectin-sense primer:5′-AAGGGTCAGGATGCTACTGTT-3′ | This paper | N/A |
Mouse adiponectin-antisense primer:5′-AGTAACGTCATCTTCGGCATGA-3′ | This paper | N/A |
Mouse ATF3-sense primer:5′-CTCCTGGGTCACTGGTATTTG-3′ | This paper | N/A |
Mouse ATF3-antisense primer:5′-CCGATGGCAGAGGTGTTTAT-3′ | This paper | N/A |
Mouse GAPDH-sense primer:5′-GGAGC CAAACGGGTCATCATCTC-3′ | This paper | N/A |
Mouse GAPDH-antisense primer:5′-GAGG GGCCATCCACAGTCTTCT-3′ | This paper | N/A |
Mouse MCP-1-sense primer:5′-ACTGAAGCTCGTACTCTC-3′ | This paper | N/A |
Mouse MCP-1-antisense primer:5′-CTTGGGTTGTGGAGTGAG-3′ | This paper | N/A |
Recombinant DNA | ||
AAV8-ATF3 | Cheng et al., 201916 | N/A |
AAV8-GFP | Cheng et al., 201916 | N/A |
Software and algorithms | ||
ImageJ | Schneider et al., 201223 | https://imagej.nih.gov/ij/ |
statistical software Prism 8.0 | GraphPad Software | https://www.graphpad.com/scientific-software/prism/ |
Other | ||
HFD (60% kcal from fat) | Research Diets Inc. | Cat# D12492 |
Resource availability
Lead contact
Further information and requests for animal resources, reagents, and data should be directed to and will be fulfilled by the lead contact, Tzu-Ling Tseng ([email protected]).
Materials availability
This study did not generate new unique reagents.
Data and code availability
All data reported in this paper will be shared by the lead contact, Tzu-Ling Tseng upon request.
This paper does not report the original code.
Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.
Experimental model and subject details
Animals and general procedures
Female ATF3-KO mice were kindly provided by Dr. Tsonwin Hai as previously described.15 Six-week-old male C57BL/6 mice, weighing 20–25 g, were purchased from BioLASCO Co., Ltd. Taipei, Taiwan and were used for crossbreeding. The female ATF3−/− mice allele was backcrossed into male C57BL/6 mice for at least seven generations before the experiments. The mice were housed at the Animal Center of Tzu Chi University. All mice were fed standard conditional chow before applying the different feeding formulas.16 All experimental procedures and protocols were performed according to the 3Rs guidelines and their relevant documents and were approved by the Animal Care and Use Committee of Tzu Chi Hospital in Taiwan (approval no.: 108–39 and 110–47).
The high-fat mouse model used in the present study was based on previous studies showing that ATF3 regulates adipocyte browning and resistance to obesity in mice.16 All animal care and experimental procedures were performed using standard laboratory procedures. Six-week-old male wild-type (WT) and ATF3−/− mice were fed a chow diet (normal diet [ND]; 4% kcal from fat) or HFD (60% kcal from fat), and their food intake was recorded weekly. Body weight was measured every week throughout the experiments.16
Method details
Non-invasive blood pressure detection
The non-invasive blood pressure detection procedure follows the original instructions from the BP-2000 Blood Pressure Analysis System (Visitech BP-2000, Visitech Systems, North Carolina, USA). During a measurement period, the experimental mice were conscious and did not require any anesthesia. The experimental animals were required to adapt for one month to cuff pressure response. The acclimatization experiments were conducted three times a week simultaneously and in a quiet space before elevated blood pressure and heartbeat measurements. At least five preliminary recordings were made in each experiment to allow the animal to warm up sufficiently to generate good blood flow to the tail. The tail must be on an excellent pulse signal at the bottom of the sensor’s V-groove. To do this, the tail-cuff handle was pushed into the grommet where the blue cuff tube goes through, and the distal tail was taped to the flat part of the sensor (the front closest to the platform). The pulse shape was checked during the detection phase to verify that the pulse amplitude was large enough. If the amplitude was too small, the temperature was set to at least 36°C. When the animal was stable measurements were taken and the measurement period did not exceed 30 min. Diastolic and systolic blood pressure were determined by monitoring vasodilation as the occlusive cuff expanded.44
Glucose tolerance testing
In the testing period, mice were fasted overnight for 16 h before receiving an intraperitoneal (IP) administration of 1.5 g glucose kg−1 body weight in saline. Plasma glucose levels were measured from the tail blood at 0, 15, 30, 45, 60, and 120 min after glucose injection. All the mice were sacrificed after initial anesthesia with isoflurane (2%) for 5 min and the deep induction of anesthesia with 0.5 g/kg urethane (intraperitoneal injection) for 10 min. The selected arteries were dissected and collected for further analysis. Whole blood samples (1 mL) were centrifuged at 11,269 × g per min for 10 min at 4°C, and plasma samples were frozen at ˗80°C for subsequent assays to measure cytokine levels.16
Production of recombinant adeno-associated virus (AAV) for ATF3 gene function restoring
This procedure has been described previously.16 Full-length ATF3 was obtained by the polymerase chain reaction (PCR) amplification of a human complementary cDNA library and cloned into the XbaI/HindIII sites of a pAAV-MCS vector. A three-plasmid co-transfection method was used to produce the AAV. The plasmids used for transfection included the AAV-CMV-ATF3 plasmid with gene expression driven by the cytomegalovirus promoter, which carried the promoter-driven transgene flanked by AAV inverted terminal repeats; the helper plasmid, which contained helper genes from the adenovirus; and the pseudotyped AAV packaging plasmid containing the AAV8 serotype capsid gene coupled with the AAV2 rep gene. AAV8-green fluorescent protein (GFP; control) and AAV8-ATF3 (experimental group) were purified twice by cesium chloride gradient ultracentrifugation. The titers of the vector genome particles were determined as previously described.16 Recombinant viruses with 1 × 1012 viral particles in 30 μL of PBS (PBS) were injected into a mouse tail vein after 6 and 8 weeks of HFD feeding (Figures S5, S6 and S7).
PVAT, BAT, and iWAT protein extraction
The adipocyte extract methods were performed and modified by published reports.29,45 Total protein was extracted from 22-week-old ATF3−/− and wild-type control mice; the thoracic aortic PVAT, suprascapular BAT, and inguinal WAT (50 mg) were dissected and washed three times in ice-cold PBS (PBS) solution (10 mM Na2HPO4, 10 mM H2PO4, 0.9 g NaCl/100 mL, pH 7.4) to remove blood in the tissue. Next, the PVAT was homogenized in ice-cold protein extract with lysis buffer (500 μL of Krebs’ solution with 20 μL of protease inhibitor cocktail; 1 mL/50 mg of vol/wt tissue) using a plastic homogenizer ten times on the ice. After vortexing, the sonication process was carried out at a frequency of 40 kHz at 37°C for 90 min. This sonication method increases the quality of adipocyte substances extracted and ensures active substances’ stability based on cavitation theory.45 The experiment was performed at a constant temperature of 35 ± 1°C. The resulting extracts were centrifuged at 3,000 × g at 4°C for 10 min to eliminate lipids and debris, and the supernatants were transferred to tubes. Protein concentrations were quantified using bicinchoninic acid (BCA) protein assay reagent (Pierce Chemicals, Rockford, IL, USA). All protein extracts from thoracic aortic PVAT, suprascapular BAT, and inguinal WAT (stock concentration, 500 μg/mL) were dissolved in pre-warmed Krebs’ solution (working concentration, 50 μg/mL) and then added and incubated (final concentration in organ bath, 5 μg/mL).29
In vitro blood vessel myography for evaluating vascular tension reactivity
The reactivity of the isolated arterial ring segments (length, 4 mm) was assessed using blood vessel myography, as previously described.46 The thoracic aortic ring segment was mounted on stainless steel rods in a tissue bath containing 10 mL Krebs’ solution at 37°C and equilibrated with 95% O2 and 5% CO2. Tension changes in the thoracic aortic ring were measured by using an isometric transducer (FT03C; Grass Technology, West Warwick, RI, USA) and recorded on a PowerLab data acquisition system (ADInstruments Pty Ltd, NSW, Australia). A simple scheme of the in vitro blood vessel myography system and original pictures of thoracic aortic ring preparation (Supplementary information, Figure S1). Experimental design for concentration curve study: The presence of functional endothelial cells (ECs) was verified by evaluating the acetylcholine (1 μM)-induced concentration-dependent relaxation of arteries pre-contracted with phenylephrine (PhE; 3 μM). After washing with Krebs’ solution under active muscle tone-induced vasoconstriction in a concentration-dependent manner with different agents, the segments were induced with potassium chloride (KCl; 7.5–120 mM), thromboxane A2 analog U46619 (0.1 nM‒1 μM), the α/β-receptor agonist NE (0.1 nM‒1 μM), or 5-HT2A receptor agonist serotonin (5-HT; 1 nM‒3 μM) in the organ bath. Experimental design for different region adipose tissue extraction experiments combined with blood vessel myography: Following standard procedures for in vitro blood vessel myography as previously described, after verifying that the endothelium was functional based on ACh (1 μM)-induced relaxation of the arteries pre-contracted with PE (1 μM), the samples were washed with pre-warmed Krebs’ solution. The adipose tissue extracts from thoracic aortic PVAT, suprascapular BAT, and inguinal WAT (stock concentration, 500 μg/mL) were dissolved in pre-warmed Krebs’ solution (working concentration, 50 μg/mL) and then added and incubated (final concentration in organ bath, 5 μg/mL) for 15 min for recording. Krebs’ solution (as a control solvent) alone was used as a negative control. After wash, a similar amount of active muscle tone was induced again with PE (1 μM). In the presence of PE-induced active muscle tone, adipose tissue extracts were applied to the organ bath to induce a relaxation response for 15 min. After washing with Krebs’ solution under the active muscle tone, which was re-induced with PhE (1 μM), the NO donor sodium nitroprusside (SNP; 0.1 nM to 100 μM) was added to induce the maximal relaxation of the arteries. The response to treatment with 120 mM KCl was considered to define 100% contraction and was used to determine the percent changes in contraction observed in response to other treatments.47
Determination of MCP-1, C-reactive protein, IL-6, resistin, serpin E1, and adiponectin levels in PVAT and circulating blood samples
Adipokine production in thoracic aortic PVAT tissues was analyzed using the Proteome Profiler Mouse Adipokine Array Kit (R&D Systems, Minneapolis, MN, USA), according to standard procedures.16 Blots were developed using enhanced chemiluminescence (ECL) and a UVP ChemStudio PLUS Imaging System (Analytik Jena, Jena, Germany). Individual bands were quantified using the ImageJ software (National Institute of Mental Health). Plasma adipokine concentrations were measured by an enzyme-linked immunosorbent assay (ELISA), according to previously published procedures.16,47 The ELISA kits for mouse adiponectin (MyBioSource, San Diego, CA, USA) and monocyte chemoattractant protein (MCP)-1 were obtained from Enzo Life Sciences (Lausen, Switzerland). The absorbance of the samples was measured at 450 nm, and the plasma concentrations of the adipokines were calculated and compared to the standard curves.16,47
Histopathology
A standard histopathological examination of the thoracic aortic specimens was performed.47 In addition, 2-μm-thick paraffin-embedded cross-sections of aortic specimens were stained with hematoxylin and eosin Y dye. The stained slides were examined under a light microscope (BX43 model, Olympus, Tokyo, Japan). For objective comparisons, five sections from each tissue block of five mice were examined, and assessments were conducted in a single-blinded fashion. The inner diameter, media area, adventitia area, and lipid size (scale range: <20 μm, 20–40 μm, >40 μm) measurements were analyzed using the ImageJ software (National Institute of Mental Health).28 The adventitia to media (A/M) ratio was calculated using the media plus adventitial layers in relation to the total area of the arterial wall.48
Immunofluorescence
Standard immunofluorescence techniques were used to assess the thoracic aortic specimens.47 In brief, 2-μm-thick paraffin-embedded aortic tissue sections were processed by antigen retrieval, cell membrane permeabilization with 0.1% Triton X-100, and blocking non-specific interactions with hydrogen peroxide (Thermo Scientific, MA, USA). Sections were washed before being incubated separately with the following antibodies: goat polyclonal anti-MCP-1 antibody (1:50; Santa Cruz Biotechnology, TX, USA), rabbit monoclonal anti-adiponectin antibody (1:200; Invitrogen, Carlsbad, California, USA), rabbit polyclonal anti-adiponectin receptor-1 (AdipoR1) antibody (1:200; GeneTex, Irvine, CA, USA), and mouse monoclonal anti-SM22 antibody (1:200; Millipore, Billerica, MA, USA). The following secondary antibodies were used: Hilyte Fluor 488-labeled anti-goat IgG for MCP-1 (1:200; AnaSpec, Fremont, CA, USA), Hilyte Fluor 488-labeled anti-rabbit IgG for AdipoR1 (1:200; AnaSpec), Hilyte Fluor 555-labeled anti-rabbit IgG for adiponectin (1:200; AnaSpec), and Hilyte Fluor 555-labeled anti-mouse IgG for SM22 (1:200; AnaSpec). For the negative controls, various sections were incubated with secondary antibodies alone, and 4ʹ,6-diamidino-2ʹ-phenylindole (DAPI) was used to counter stain the nuclei (1:200; KPL, Washington DC, USA). All sections were examined using an Olympus BX43 fluorescence microscope affixed to a DP-72 digital camera system (Olympus, Center Valley, PA, USA). Tissue blocks from all the mice were evaluated for statistical analyses. The intensities of the orange and green fluorescent signals were analyzed using the ImageJ software (National Institute of Mental Health).47
Quantification of the immunofluorescent intensity
The method for the standardized relative quantification of immunofluorescence (I) staining of tissue sections was performed according to Arques et al. (https://doi.org/10.1038/protex.2012.008). In brief, fluorescent images were defined in regions of interest (ROI) or randomly selected areas using polygon and plot z axis profile selection tools, set standard quantification sizes, and delimited fluorescence from PVAT and vascular smooth muscle. For MCP-1, adiponectin-I, or adipoR1-I, the selected images were imported into the red-green-blue (RGB) channel and measured using the ImageJ ROI manager to assess the gray integrated density value (IDV). The fluorescence intensity of individual SM22-immunoreactive smooth muscle cells was also determined. This was followed by placing horizontal and vertical lines of various orientations and lengths across the smooth muscle layers or PVAT sections. The intensities of the gray scale values (pixels) at each point along the line were measured. Thus, changes in the intensities related to immunoreactivity across the width of each muscle layer were determined. On average, two lines were drawn on each layer. Similarly, the selected images were imported to the RGB channel and measured by the ROI manager to assess the gray IDV.47
Immunoblotting
Conventional procedures for isolating cytoplasmic and membrane proteins and electrophoresis with western blotting were performed as previously described.47 The primary antibodies included rabbit polyclonal anti-MCP-1 antibody (1:500; Arigo, Hsinchu City, Taiwan), rabbit monoclonal anti-adiponectin (1:2000; Invitrogen, Carlsbad, California, USA), and rabbit monoclonal anti-AdipoR1 (1:2000; GeneTex, Irvine, CA, USA). Horseradish peroxidase-conjugated anti-mouse and anti-rabbit IgG secondary antibodies (1:2000; GeneTex) with an ECL horseradish peroxidase substrate detection kit (Millipore, MA, USA) were used to visualize the bands. The membranes were also incubated with a mouse monoclonal anti-actin antibody (1:4000; Thermo Fisher Scientific, IL, USA) for normalization and to determine equal protein loading. Individual bands were quantified using ImageJ (National Institute of Mental Health).23
Real-time PCR
Total RNA was extracted from thoracic aortic PVAT using TRIzol Reagent (Invitrogen). The RNA was reverse transcribed to cDNA using the iScript cDNA Synthesis Kit (Bio-Rad, Hercules, CA, USA). Real-time qPCR analysis was performed using the ABI PRISM 7700 Sequence Detection System (Applied Biosystems, Grand Island, NY, USA) with SYBR green (Bio-Rad).16,32 The PCR primers used were as follows:
Mouse adiponectin-sense primer: 5′-AAGGGTCAGGATGCTACTGTT-3′,
Mouse adiponectin-antisense primer: 5′-AGTAACGTCATCTTCGGCATGA-3′,
Mouse ATF3-sense primer: 5′-CTCCTGGGTCACTGGTATTTG-3′,
Mouse ATF3-antisense primer: 5′-CCGATGGCAGAGGTGTTTAT-3′,
Mouse GAPDH-sense primer: 5′-GGAGCCAAACGGGTCATCATCTC-3′,
Mouse GAPDH-antisense primer: 5′-GAGGGGCCATCCACAGTCTTCT-3′,
Mouse MCP-1-sense primer: 5′-ACTGAAGCTCGTACTCTC-3′,
Mouse MCP-1-antisense primer: 5′-CTTGGGTTGTGGAGTGAG-3′.
Microarray data sources
The microarray experimental data shown in Figure 2 were obtained from the Gene Expression Omnibus (GEO; https://www.ncbi.nlm.nih.gov/geo/) of the NCBI. These included data from a mouse model of hypertension (accession no. GDS3672, aorta from hypertensive and normotensive mice; accession no. GDS3675, kidneys from hypertensive and normotensive mice; accession no. GDS3673, hearts from hypertensive and normotensive mice; accession no. GDS3674, liver from hypertensive and normotensive mice)27 and human subjects (accession no. GSE24752, peripheral blood cells from healthy and hypertensive individuals).26
Drugs
SNP, Tween 20, Triton X-100, absolute ethanol, methanol, PhE, KCl, NE, 5-HT, U46619, hematoxylin, and eosin Y were purchased from Sigma Chemicals (Chicago, IL, USA).
Quantification and statistical analysis
Data are presented as the mean ± SEM. 42 All data were analyzed by using the statistical software Prism 8.0 (GraphPad Software, San Diego, CA, USA). Statistical comparisons between two groups were performed using unpaired t-test and for more than two groups using one-way ANOVA. Statistical comparisons for different phenotype groups, time-course, and concentration-induced vascular reactivity were performed using two-way ANOVA. All concentration curves were analyzed using the generalized estimating equation (GEE) method. Post hoc comparisons between the groups were performed using the Bonferroni multiple comparison test. A p value < 0.05 was considered statistically significant. A correlation analysis was performed using Pearson’s correlation coefficient (Figures 6F and S6E).
Acknowledgments
The authors acknowledge the core facilities of the Advanced Instrumentation Center of the Department of Medicine Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Function, Hualien, Taiwan. The authors thank Editage (https://www.editage.com.tw) for English language editing. Funding: this work was supported by the Ministry of Science and Technology [MOST 108-2314-B-038-048-MY3 to H Lin; MOST 110-2320-B-303-001-MY3 and MOST 111-2320-B-303-001 to TL Tseng], Buddhist Tzu Chi General Hospital Grant TCMMP (109-1) Taiwan to TL Tseng.
Author contributions
Conceptualization, T.L.T., and H.L.; Methodology, H.F.L., H.T.L., P.Y.C., and T.L.T.; Investigation, H.F.L., H.T.L., P.Y.C., and T.L.T.; Validation, T.L.T., and H.L.; Writing—Original Draft, T.L.T., and H.L.; Writing—Review & Editing, T.L.T., and H.L.; Project Administration, T.L.T., and H.L.
Declaration of interests
The authors declare no competing interests.
Footnotes
Supplemental information can be found online at https://doi.org/10.1016/j.isci.2022.105631.
References
Articles from iScience are provided here courtesy of Elsevier
Full text links
Read article at publisher's site: https://doi.org/10.1016/j.isci.2022.105631
Read article for free, from open access legal sources, via Unpaywall: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9707070
Citations & impact
Impact metrics
Citations of article over time
Alternative metrics
Discover the attention surrounding your research
https://www.altmetric.com/details/139587901
Smart citations by scite.ai
Explore citation contexts and check if this article has been
supported or disputed.
https://scite.ai/reports/10.1016/j.isci.2022.105631
Article citations
Cellular, Molecular and Clinical Aspects of Aortic Aneurysm-Vascular Physiology and Pathophysiology.
Cells, 13(3):274, 01 Feb 2024
Cited by: 1 article | PMID: 38334666 | PMCID: PMC10854611
Review Free full text in Europe PMC
Perivascular adipose tissue and adipocyte-derived exosomal miRNAs maintain vascular homeostasis.
Heliyon, 9(12):e22607, 20 Nov 2023
Cited by: 0 articles | PMID: 38076178 | PMCID: PMC10709399
Review Free full text in Europe PMC
Fat and inflammation: adipocyte-myeloid cell crosstalk in atherosclerosis.
Front Immunol, 14:1238664, 15 Sep 2023
Cited by: 0 articles | PMID: 37781401 | PMCID: PMC10540690
Review Free full text in Europe PMC
The Association between Serum Adiponectin Levels and Endothelial Function in Non-Dialysis-Dependent Chronic Kidney Disease Patients.
Biomedicines, 11(8):2174, 02 Aug 2023
Cited by: 1 article | PMID: 37626670 | PMCID: PMC10452815
Kidney lipid dysmetabolism and lipid droplet accumulation in chronic kidney disease.
Nat Rev Nephrol, 19(10):629-645, 27 Jul 2023
Cited by: 35 articles | PMID: 37500941
Review
Go to all (6) article citations
Data
Data behind the article
This data has been text mined from the article, or deposited into data resources.
GEO - Gene Expression Omnibus (5)
- (2 citations) GEO - GDS3673
- (2 citations) GEO - GDS3672
- (2 citations) GEO - GSE24752
- (2 citations) GEO - GDS3675
- (2 citations) GEO - GDS3674
Nucleotide Sequences
- (1 citation) ENA - D12492
Similar Articles
To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.
miRNA-22 is involved in the aortic reactivity in physiological conditions and mediates obesity-induced perivascular adipose tissue dysfunction.
Life Sci, 316:121416, 21 Jan 2023
Cited by: 1 article | PMID: 36690245
Dahl SS rats demonstrate enhanced aortic perivascular adipose tissue-mediated buffering of vasoconstriction through activation of NOS in the endothelium.
Am J Physiol Regul Integr Comp Physiol, 310(3):R286-96, 25 Nov 2015
Cited by: 11 articles | PMID: 26608658 | PMCID: PMC4796750
Loss of perivascular adipose tissue on peroxisome proliferator-activated receptor-γ deletion in smooth muscle cells impairs intravascular thermoregulation and enhances atherosclerosis.
Circulation, 126(9):1067-1078, 01 Aug 2012
Cited by: 202 articles | PMID: 22855570 | PMCID: PMC3493564
PVAT and Its Relation to Brown, Beige, and White Adipose Tissue in Development and Function.
Front Physiol, 9:70, 06 Feb 2018
Cited by: 57 articles | PMID: 29467675 | PMCID: PMC5808192
Review Free full text in Europe PMC
Funding
Funders who supported this work.