Gut Microbiota-Related Evidence Provides New Insights Into the Association Between Activating Transcription Factor 4 and Development of Salt-Induced Hypertension in Mice

Tian-Hao Liu, Wen-Cong Tao, Qiu-Er Liang, Wan-Qing Tu, Ya Xiao, Li-Guo Chen, Tian-Hao Liu, Wen-Cong Tao, Qiu-Er Liang, Wan-Qing Tu, Ya Xiao, Li-Guo Chen

Abstract

Activating transcription factor 4 (ATF4), which regulates genes associated with endoplasmic reticulum stress, apoptosis, autophagy, the gut microbiome, and metabolism, has been implicated in many diseases. However, its mechanistic role in hypertension remains unclear. In the present study, we investigated its role in salt-sensitive hypertensive mice. Wild-type (WT) C57BL/6J mice were used to establish Atf4 knockout (KO) and overexpression mice using CRISPR-Cas9 and lentiviral overexpression vectors. Then, fecal microbiota transplantation (FMT) from Atf4 ± mice and vitamin K2 (VK2) supplementation were separately carried out in high-salt-diet (8% NaCl)-induced mice for 4 weeks. We found that Atf4 KO inhibited and Atf4 overexpression enhanced the increase in blood pressure and endothelial dysfunction induced by high salt intake in mice, while regulating the gut microbiota composition and VK2 expression. It was further verified that ATF4 is involved in the regulation of salt-sensitive hypertension and vascular endothelial function, which is achieved through association with gut microbiota and may be related to VK2 and different bacteria such as Dubosiella. In addition, we found that VK2 supplementation prevents the development of salt-sensitive hypertension and maintains vascular endothelial function; moreover, VK2 supplementation increases the abundance of intestinal Dubosiella and downregulates the relative expression of Atf4 in the thoracic aorta of mice. We conclude that ATF4 plays an important role in regulating gut microbiota and VK2 production, providing new insights into the association between ATF4 and development of salt-induced hypertension in mice, meanwhile contributing to the development for a new preventive strategy of hypertension.

Keywords: gene regulation; gut microbiota; hypertension; mice; vitamin K2.

Copyright © 2020 Liu, Tao, Liang, Tu, Xiao and Chen.

Figures

FIGURE 1
FIGURE 1
The cartoon of experimental design to investigate the potential mechanism of ATF4 participating in high-salt diet-induced hypertension. Wild-type (WT) C57BL/6J mice, Atf4 knockout (Atf4±) and Atf4 overexpression mice were used and high-salt-diet (8% NaCl) for 4 weeks was used to induce hypertensive mice. Then, fecal microbiota transplantation from Atf4± mice and vitamin K2 supplementation were separately carried out to verify the gut microbiota-related evidence of the association between ATF4 and blood pressure. ATF4, activating transcription factor 4; KO, Atf4± mice; WT, wild-type mice; NO, nitric oxide; VEGF, vascular endothelial growth factor; ANGI, angiotensin I; ET-1, endothelin-1; KO-ND, Atf4± mice with natural diet; KO-HSD, Atf4± mice with high-salt diet; ND, WT mice with natural diet; HSD, WT mice with high-salt diet; OE, Atf4 overexpression with high-salt diet; OC, Atf4 overexpression control with high-salt diet; FMT, fecal microbiota transplantation; VK2, vitamin K2.
FIGURE 2
FIGURE 2
ATF4 altered SBP and endothelial function-related factors in high-salt-induced hypertensive mice. (A) ATF4 altered SBP in high-salt-induced hypertensive mice. ND vs HSD, HSD vs OE, OE vs OC, *p < 0.05 and **p < 0.01. (B) ATF4 altered the expression of NO in high-salt-induced hypertensive mice. (C) ATF4 altered the expression of VEGF in high-salt-induced hypertensive mice. (D) ATF4 altered the expression of ANGI in high-salt-induced hypertensive mice. (E) ATF4 altered the expression of ET-1 in high-salt-induced hypertensive mice. Data are presented as mean ± SD; ns, no significance, *p < 0.05 and **p < 0.01, n = 6–8; statistical comparisons were performed using Student t-test or one-way analysis of variance (ANOVA). ATF4, activating transcription factor 4; KO, Atf4± mice; WT, wild-type mice; SBP, systolic blood pressure; NO, nitric oxide; VEGF, vascular endothelial growth factor; ANGI, angiotensin I; ET-1, endothelin-1; KO-ND, Atf4± mice with natural diet; KO-HSD, Atf4± mice with high-salt diet; ND, WT mice with natural diet; HSD, WT mice with high-salt diet; OE, Atf4 overexpression with high-salt diet; OC, Atf4 overexpression control with high-salt diet.
FIGURE 3
FIGURE 3
ATF4 altered the composition of gut microbiota and expression levels of the microbial metabolite VK2 in high-salt-induced hypertensive mice. (A) Relative abundance plots displaying the differences in the microbial community structure at the phylum level. (B) Relative abundance plots displaying the differences in the general microbial community structure. (C) PCA plots were used to visualize differences in weighted UniFrac distances of samples of OTUs from different groups (R2 = 0.3264, p = 0.001). (D) ATF4 altered the expression of VK2. Data are presented as mean ± SD; **p < 0.01, n = 6–8; statistical comparisons were performed using Student t test or one-way analysis of variance (ANOVA). ATF4, activating transcription factor 4; KO, Atf4± mice; WT, wild-type mice; SBP, systolic blood pressure; NO, nitric oxide; VEGF, vascular endothelial growth factor; ANGI, angiotensin I; ET-1, endothelin-1; KO-ND, Atf4± mice with natural diet; KO-HSD, Atf4± mice with high-salt diet; ND, WT mice with natural diet; HSD, WT mice with high-salt diet; OE, Atf4 overexpression with high-salt diet; OC, Atf4 overexpression control with high-salt diet.
FIGURE 4
FIGURE 4
Microbial correlation in high-salt-induced hypertensive mice. (A) Spearman correlation analysis between top 20 genera and environmental factors. Red represents positive correlation, whereas blue represents negative correlation; the darker the color, the stronger the correlation. ∗0.01 < p ≤ 0.05, ∗∗0.001 < p ≤ 0.01, ∗∗∗p ≤ 0.001. (B) RDA analysis of environmental factors. The angle between the arrows of different environmental factors represents positive and negative correlations (acute angle: positive correlation; obtuse angle: negative correlation; right angle: no correlation). ATF4, activating transcription factor 4; KO, Atf4± mice; WT, wild-type mice; SBP, systolic blood pressure; NO, nitric oxide; VEGF, vascular endothelial growth factor; ANGI, angiotensin I; ET-1, endothelin-1; KO-ND, Atf4± mice with natural diet; KO-HSD, Atf4± mice with high-salt diet; ND, WT mice with natural diet; HSD, WT mice with high-salt diet; OE, Atf4 overexpression with high-salt diet; OC, Atf4 overexpression control with high-salt diet.
FIGURE 5
FIGURE 5
FMT using Atf4± mice fecal microbiota improved SBP and altered expression of endothelial function-related factors in high-salt-induced hypertensive mice. (A) FMT maintained SBP. (B) FMT upregulated the expression of NO. (C) FMT downregulated the expression of VEGF. (D) FMT downregulated the expression of ANGI. (E) FMT downregulated the expression of ET-1. (F) Examination of endothelial morphology by transmission electronic microscope (TEM). IEL, internal elastic lamina; N, nucleus; M, mitochondrion; RER, rough endoplasmic reticulum; TJ, tight junction; AP, autophagy. Data are presented as mean ± SD; **p < 0.01, n = 6–8; statistical comparisons were performed using Student t-test or one-way analysis of variance (ANOVA). FMT, fecal microbiota transplantation; SBP, systolic blood pressure; NO, nitric oxide; VEGF, vascular endothelial growth factor; ANGI, angiotensin I; ET-1, endothelin-1; ND, WT mice with natural diet; HSD, WT mice with high-salt diet.
FIGURE 6
FIGURE 6
FMT using Atf4± mice fecal microbiota altered the composition of gut microbiota and expression of the microbial metabolite VK2 in high-salt-induced hypertensive mice. (A) Venn diagram of OTUs. Different colors represent different groups. (B) Relative abundance plots displaying the differences in the microbial community structure at the phylum level. (C) Relative abundance plots displaying the differences in the general microbial community structure. (D) NMDS plots were used to visualize differences in weighted UniFrac distances at the genus level of samples from different groups. (E) The 11 taxonomic groups associated with FMT. *p < 0.05 and **p < 0.01. (F) FMT upregulated the expression of VK2. Data are presented as mean ± SD; **p < 0.01, n = 6–8; statistical comparisons were performed using Student t-test or one-way analysis of variance (ANOVA). FMT, fecal microbiota transplantation; VK2, vitamin K2; KO-ND, Atf4± mice with natural diet; ND, WT mice with natural diet; HSD, WT mice with high-salt diet.
FIGURE 7
FIGURE 7
VK2 maintained SBP and altered endothelial function-related factors in high-salt-induced hypertensive mice. (A) VK2 altered SBP. (B) VK2 upregulated the expression of NO. (C) VK2 downregulated the expression of VEGF. (D) VK2 downregulated the expression of ANGI. (E) VK2 downregulated the expression of ET-1. Data are presented as mean ± SD; *p < 0.05 and **p < 0.01, n = 6–8; statistical comparisons were performed using Student t-test or one-way analysis of variance (ANOVA). SBP, systolic blood pressure; VK2, vitamin K2; NO, nitric oxide; VEGF, vascular endothelial growth factor; ANGI, angiotensin I; ET-1, endothelin-1; ND, WT mice with natural diet; HSD, WT mice with high-salt diet.

References

    1. Bai Y., Wang S., Wang X., Weng Y., Fan X. (2019). The flavonoid-rich Quzhou Fructus Aurantii extract modulates gut microbiota and prevents obesity in high-fat diet-fed mice. Nutr. Diabetes 9 1–11. 10.1038/s41387-019-0097-6
    1. Bárcena C., Valdés-Mas R., Mayoral P., Garabaya C., Durand S. (2019). Healthspan and lifespan extension by fecal microbiota transplantation into progeroid mice. Nat. Med. 25 1234–1242. 10.1038/s41591-019-0504-5
    1. Bentley R., Meganathan R. (1982). Biosynthesis of vitamin k (Menaquinone) in bacteria. Microbiol. Rev. 3 241–280. 10.1007/BF02010678
    1. Bhalerao S., Clandinin T. R. (2012). Vitamin k2 takes charge. Science 336 1241–1242. 10.1126/science.1223812
    1. Bier A., Braun T., Khasbab R., Di Segni A., Grossman E., Haberman Y., et al. (2018). A high salt diet modulates the gut microbiota and short chain fatty acids production in a Salt-Sensitive hypertension rat model. Nutrients 10:1154. 10.3390/nu10091154
    1. Binder N. K., Brownfoot F. C., Beard S., Cannon P., Nguyen T. V., Tong S., et al. (2020). Esomeprazole and sulfasalazine in combination additively reduce sFlt-1 secretion and diminish endothelial dysfunction: Potential for a combination treatment for preeclampsia. Preg. Hyperten. 22 86–92. 10.1016/j.preghy.2020.07.013
    1. Chu H., Khosravi A., Kusumawardhani I., Kwon A., Vasconcelos A., Cunha L., et al. (2016). Gene-microbiota interactions contribute to the pathogenesis of inflammatory bowel disease. Science 352 1116–1120. 10.1126/science.aad9948
    1. Cornejo V. H., Pihán P., Vidal R. L., Hetz C. (2013). Role of the unfolded protein response in organ physiology: Lessons from mouse models. IUBMB Life 65 962–975. 10.1002/iub.1224
    1. Fastrès A., Roels E., Vangrinsven E., Taminiau B., Jabri H. (2020). Assessment of the lung microbiota in dogs: Influence of the type of breed, living conditions and canine idiopathic pulmonary fibrosis. BMC Microbiol. 20:84. 10.1186/s12866-020-01784-w
    1. Faulkner J. L., Harwood D., Bender L., Shrestha L., Brands M. W., Morwitzer M. J., et al. (2018). Lack of suppression of aldosterone production leads to Salt-Sensitive hypertension in female but not male Balb/C mice. Hypertension 72 1397–1406. 10.1161/HYPERTENSIONAHA.118.11303
    1. Haugsgjerd T. R., Egeland G. M., Nygård O. K., Vinknes K. J., Sulo G. (2020). Association of dietary vitamin K and risk of coronary heart disease in middle-age adults: The Hordaland Health Study Cohort. BMJ Open 10:e35953. 10.1136/bmjopen-2019-035953
    1. He L., Fang M., Chen L., Zhou J., Yuan J., Jing X., et al. (2016a). Transcriptome analysis of blood stasis syndrome in subjects with hypertension. J. Trad. Chin. Med. 36 173–180. 10.1016/s0254-6272(16)30024-3
    1. He L., Yuan J., Xu Q., Chen R., Chen L., Fang M., et al. (2016b). MiRNA-1283 regulates the PERK/ATF4 pathway in vascular injury by targeting ATF4. PLoS One 11:e159171. 10.1371/journal.pone.0159171
    1. Hendrik B., András B., Mina Y., Susanne H., Lajos M., Sascha H., et al. (2019). Short-Chain fatty acid propionate protects from hypertensive cardiovascular damage. Circulation 139 1407–1421. 10.1161/CIRCULATIONAHA.118.036652
    1. Hodin C. M., Verdam F. J., Grootjans J., Rensen S. S., Verheyen F. K., Dejong C. H. C., et al. (2011). Reduced Paneth cell antimicrobial protein levels correlate with activation of the unfolded protein response in the gut of obese individuals. J. Pathol. 225 276–284. 10.1002/path.2917
    1. Hu X., Deng J., Yu T., Chen S., Ge Y., Zhou Z., et al. (2019). ATF4 deficiency promotes intestinal inflammation in mice by reducing uptake of glutamine and expression of antimicrobial peptides. Gastroenterology 156 1098–1111. 10.1053/j.gastro.2018.11.033
    1. Huang Z., Ruan H., Xian L., Chen W., Jiang S., Song A., et al. (2014). The stem cell factor/Kit signalling pathway regulates mitochondrial function and energy expenditure. Nat. Commun. 5:4282. 10.1038/ncomms5282
    1. Jing L., Fangqing Z., Yidan W., Junru C., Jie T., Liu L., et al. (2017). Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome 5:14. 10.1186/s40168-016-0222-x
    1. Kurtz T. W., DiCarlo S. E., Pravenec M., Morris R. C. (2016). Vasodysfunction that involves renal vasodysfunction, not abnormally increased renal retention of sodium, accounts for the initiation of Salt-Induced hypertension. Circulation 133 881–893. 10.1161/CIRCULATIONAHA.115.017923
    1. Liao X., Song L., Zeng B., Liu B., Qiu Y., Qu H., et al. (2019). Alteration of gut microbiota induced by DPP-4i treatment improves glucose homeostasis. EBioMed. 44 665–674. 10.1016/j.ebiom.2019.03.057
    1. Liu T., Yang Z., Zhang X., Han N., Yuan J., Cheng Y. (2017). 16S rDNA analysis of the effect of fecal microbiota transplantation on pulmonary and intestinal flora. 3 Biotech 7:370. 10.1007/s13205-017-0997-x
    1. Mansour A. G., Ahdab R., Daaboul Y., Korjian S., Morrison D. A., Hariri E., et al. (2019). Vitamin k2 status and arterial stiffness among untreated migraine patients: A case-control study. Headache J. Head Face Pain 60 589–599. 10.1111/head.13715
    1. Masuoka H. C., Townes T. M. (2002). Targeted disruption of the activating transcription factor 4 gene results in severe fetal anemia in mice. Blood 99 736–745. 10.1182/blood.V99.3.736
    1. Paczula A., Wiecek A., Piecha G. (2019). Cardiotonic steroids—a possible link between High-Salt diet and organ damage. Int. J. Mol. Sci. 20:590. 10.3390/ijms20030590
    1. Ponziani F. R., Pompili M., Di Stasio E., Zocco M. A., Gasbarrini A., Flore R. (2017). Subclinical atherosclerosis is linked to small intestinal bacterial overgrowth via vitamin K2-dependent mechanisms. World J. Gastroenterol. 23:1241. 10.3748/wjg.v23.i7.1241
    1. Quirós P. M., Prado M. A., Zamboni N., D’Amico D., Williams R. W., Finley D., et al. (2017). Multi-omics analysis identifies ATF4 as a key regulator of the mitochondrial stress response in mammals. J. Cell Biol. 216 2027–2045. 10.1083/jcb.201702058
    1. Ren L., Peng C., Hu X., Han Y., Huang H. (2020). Microbial production of vitamin K2: Current status and future prospects. Biotechnol. Adv. 39:107453. 10.1016/j.biotechadv.2019.107453
    1. Rufaihah A. J., Johari N. A., Vaibavi S. R., Plotkin M., Di Thien D. T., Kofidis T., et al. (2017). Dual delivery of VEGF and ANG-1 in ischemic hearts using an injectable hydrogel. Acta Biomater. 48 58–67. 10.1016/j.actbio.2016.10.013
    1. Sakai T., Kurokawa R., Hirano S., Imai J. (2019). Hydrogen indirectly suppresses increases in hydrogen peroxide in cytoplasmic hydroxyl Radical-Induced cells and suppresses cellular senescence. Int. J. Mol. Sci. 20:456. 10.3390/ijms20020456
    1. Stebegg M., Silva-Cayetano A., Innocentin S., Jenkins T. P., Cantacessi C., Gilbert C., et al. (2019). Heterochronic faecal transplantation boosts gut germinal centres in aged mice. Nat. Commun. 10:2443. 10.1038/s41467-019-10430-7
    1. Tang W. H. W., Li D. Y., Hazen S. L. (2019). Dietary metabolism, the gut microbiome, and heart failure. Nat. Rev. Cardiol. 16 137–154. 10.1038/s41569-018-0108-7
    1. Tameire F., Verginadis I. I., Leli N. M., Polte C., Conn C. S., Ojha R., et al. (2019). Author Correction: ATF4 couples MYC-dependent translational activity to bioenergetic demands during tumour progression. Nat. Cell Biol. 21:1052. 10.1038/s41556-019-0370-x
    1. Touyz R. M., Camargo L. L. (2019). Microglia, the missing link in the Brain-Gut-Hypertension axis. Circulation Res. 124 671–673. 10.1161/CIRCRESAHA.119.314718
    1. Viennois E., Chassaing B., Tahsin A., Pujada A., Wang L., Gewirtz A., et al. (2019). Host-derived fecal microRNAs can indicate gut microbiota healthiness and ability to induce inflammation. Theranostics 9 4542–4557. 10.7150/thno.35282
    1. Vos M., Esposito G., Edirisinghe J. N., Vilain S., Haddad D. M., Slabbaert J. R., et al. (2012) Vitamin k2 is a mitochondrial electron carrier that rescues pink1 deficiency. Science 336 1306–1310. 10.1126/science.1218632
    1. Waghulde H., Cheng X., Galla S., Mell B., Cai J., Pruett-Miller S. M., et al. (2018). Attenuation of microbiotal dysbiosis and hypertension in aCRISPR/Cas9 gene ablation rat model ofGPER1. Hypertension 72 1125–1132. 10.1161/HYPERTENSIONAHA.118.11175
    1. Wang J., Thingholm L., Skiecevičienė J., Rausch P., Kummen M., Hov J. R., et al. (2016). Genome-wide association analysis identifies variation in vitamin D receptor and other host factors influencing the gut microbiota. Nat. Genet. 48 1396–1406. 10.1038/ng.3695
    1. Wang W., Lian N., Li L., Moss H. E., Wang W., Perrien D. S., et al. (2009). Atf4 regulates chondrocyte proliferation and differentiation during endochondral ossification by activating Ihh transcription. Development 136 4143–4153. 10.1242/dev.043281
    1. Wang Z., Chen Z., Zhang L., Wang X., Hao G., Zhang Z., et al. (2018). Status of hypertension in china: Results from the china hypertension survey, 2012-2015. Circulation 137 2344–2356. 10.1161/CIRCULATIONAHA.117.032380
    1. Wilck N., Matus M. G., Kearney S. M., Olesen S. W., Forslund K., Bartolomaeus H., et al. (2017). Salt-responsive gut commensal modulates TH17 axis and disease. Nature 551 585–589. 10.1038/nature24628
    1. Wu X., Luo J., Liu H., Cui W., Guo W., Zhao L., et al. (2020). Recombinant adiponectin peptide promotes neuronal survival after intracerebral haemorrhage by suppressing mitochondrial and ATF4-CHOP apoptosis pathways in diabetic mice via Smad3 signalling inhibition. Cell Proliferat. 53:e12759. 10.1111/cpr.12759
    1. Xu D., Dai W., Kutzler L., Lacko H. A., Jefferson L. S., Dennis M. D., et al. (2019). ATF4-Mediated upregulation of REDD1 and sestrin2 suppresses mTORC1 activity during prolonged leucine deprivation. J. Nutr. 150 1022–1030. 10.1093/jn/nxz309
    1. Xu H. S., Duan J., Dai S., Wu Y., Sun R., Ren J. (2004). Phytoestrogen alpha-zearalanol antagonizes oxidized LDL-induced inhibition of nitric oxide production and stimulation of endothelin-1 release in human umbilical vein endothelial cells. Endocrine 25 235–245. 10.1385/ENDO:25:3:235
    1. Yan Z., Gao X., Li T., Wei B., Wang P., Yang Y., et al. (2018). Fecal microbiota transplantation in experimental ulcerative colitis reveals associated gut microbial and host metabolic reprogramming. Appl. Environ. Microbiol. 84 434–418e. 10.1128/AEM.00434-18
    1. Yang Y., Liu X., Liu Y., Fu H., Gao Y., Liu X., et al. (2018). The development of salt-sensitive hypertension regulated by PSGL-1 gene in mice. Cell Biosci. 8:20. 10.1186/s13578-018-0218-2
    1. Zhang M., Wang S., Wang Y., Zhang Y., Hao C., Harris R. C. (2018). Renal medullary interstitial COX-2 (Cyclooxygenase-2) is essential in preventing Salt-Sensitive hypertension and maintaining renal inner Medulla/Papilla structural integrity. Hypertension 72 1172–1179. 10.1161/HYPERTENSIONAHA.118.11694

Source: PubMed

3
Subscribe