Pan-HDAC (histone Deacetylase) Inhibitors Increase Susceptibility of Thoracic Aortic Aneurysm and Dissection in Mice
Shumin Zhang,Zhujiang Liu,Nan Xie,Chuanbo Huang,Zhiqing Li,Fang Yu,Yi Fu,Qinghua Cui,Wei Kong
DOI: https://doi.org/10.1161/atvbaha.121.316808
2021-01-01
Abstract:HomeArteriosclerosis, Thrombosis, and Vascular BiologyVol. 41, No. 11Pan-HDAC (Histone Deacetylase) Inhibitors Increase Susceptibility of Thoracic Aortic Aneurysm and Dissection in Mice Free AccessLetterPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyRedditDiggEmail Jump toFree AccessLetterPDF/EPUBPan-HDAC (Histone Deacetylase) Inhibitors Increase Susceptibility of Thoracic Aortic Aneurysm and Dissection in Mice Shumin Zhang, Zhujiang Liu, Nan Xie, Chuanbo Huang, Zhiqing Li, Fang Yu, Yi Fu, Qinghua Cui and Wei Kong Shumin ZhangShumin Zhang Department of Physiology and Pathophysiology (S.Z., Z. Liu, N.X., Z. Li, F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China. Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China (S.Z., Z. Liu, N.X., Z. Li, F.Y., Y.F., W.K., C.H., Q.C.). *S. Zhang, Z. Liu, and N. Xie contributed equally. Search for more papers by this author , Zhujiang LiuZhujiang Liu Department of Physiology and Pathophysiology (S.Z., Z. Liu, N.X., Z. Li, F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China. Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China (S.Z., Z. Liu, N.X., Z. Li, F.Y., Y.F., W.K., C.H., Q.C.). *S. Zhang, Z. Liu, and N. Xie contributed equally. Search for more papers by this author , Nan XieNan Xie Department of Physiology and Pathophysiology (S.Z., Z. Liu, N.X., Z. Li, F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China. Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China (S.Z., Z. Liu, N.X., Z. Li, F.Y., Y.F., W.K., C.H., Q.C.). *S. Zhang, Z. Liu, and N. Xie contributed equally. Search for more papers by this author , Chuanbo HuangChuanbo Huang Department of Biomedical Informatics, Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine (C.H., Q.C.), School of Basic Medical Sciences, Peking University, Beijing, China. Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China (S.Z., Z. Liu, N.X., Z. Li, F.Y., Y.F., W.K., C.H., Q.C.). Search for more papers by this author , Zhiqing LiZhiqing Li Department of Physiology and Pathophysiology (S.Z., Z. Liu, N.X., Z. Li, F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China. Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China (S.Z., Z. Liu, N.X., Z. Li, F.Y., Y.F., W.K., C.H., Q.C.). Search for more papers by this author , Fang YuFang Yu Department of Physiology and Pathophysiology (S.Z., Z. Liu, N.X., Z. Li, F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China. Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China (S.Z., Z. Liu, N.X., Z. Li, F.Y., Y.F., W.K., C.H., Q.C.). Search for more papers by this author , Yi FuYi Fu Department of Physiology and Pathophysiology (S.Z., Z. Liu, N.X., Z. Li, F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China. Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China (S.Z., Z. Liu, N.X., Z. Li, F.Y., Y.F., W.K., C.H., Q.C.). Search for more papers by this author , Qinghua CuiQinghua Cui Correspondence to: Wei Kong, MD, PhD, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China, Email E-mail Address: [email protected] Department of Biomedical Informatics, Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine (C.H., Q.C.), School of Basic Medical Sciences, Peking University, Beijing, China. Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China (S.Z., Z. Liu, N.X., Z. Li, F.Y., Y.F., W.K., C.H., Q.C.). Search for more papers by this author and Wei KongWei Kong Correspondence to: Wei Kong, MD, PhD, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China, Email E-mail Address: [email protected] https://orcid.org/0000-0001-6720-6810 Department of Physiology and Pathophysiology (S.Z., Z. Liu, N.X., Z. Li, F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China. Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China (S.Z., Z. Liu, N.X., Z. Li, F.Y., Y.F., W.K., C.H., Q.C.). Search for more papers by this author Originally published16 Sep 2021https://doi.org/10.1161/ATVBAHA.121.316808Arteriosclerosis, Thrombosis, and Vascular Biology. 2021;41:2848–2850Other version(s) of this articleYou are viewing the most recent version of this article. Previous versions: September 16, 2021: Ahead of Print Thoracic aortic aneurysm and dissection (TAAD) is a highly lethal aortic disease. Administration of a lysyl oxidase inhibitor, β-aminopropionitrile (BAPN) fumarate, induces medial degeneration of aorta and results in TAAD formation in mice, characterizing by local dilation of the thoracic aortic wall with a maximum external diameter of >50% compared with the adjacent intact part of the aorta, a tear in the intimal layer of the aorta, extracellular matrix degradation, and inflammation. The pathogenesis of TAAD involves both genetic mutations such as FBN1, LOX, MYH11, and COL3A1 and environmental factors including aging, hypertension, and inflammation. Of note, 2 recent clinical studies indicate that the application of fluoroquinolone antibiotics increases the risk of TAAD.1,2 The cause-effect of fluoroquinolone antibiotics to TAAD is further confirmed by supplementation of ciprofloxacin to Ang II (angiotensin II)–infused mice.3 This alarming observation has raised concern in patients with risk for developing TAAD. Whether other clinical applicable drugs also increase the risk on TAAD progression or rupture?The Connectivity Map has provided a data-driven and systematic approach for discovering associations among chemicals, genes, and diseases based on genome-wide expression profiling.4 In principle, drugs with similar gene expression signatures may share similar mechanisms, chemical and physiological processes, and efficacies for similar diseases. By using 6 different fluoroquinolone antibiotics including pefloxacin, ofloxacin, ciprofloxacin, norfloxacin, lomefloxacin, and enoxacin as baits, we found that panobinostat and cefixime exhibited high degree of similarity with fluoroquinolones based on the Spearman correlation score by the Connectivity Map sorting algorithm (Figure [A]). Panobinostat is a pan-HDAC (histone deacetylase) inhibitor clinically applied for multiple myeloma therapy, and its Spearman similarity score was 0.577. Cefixime is a third-generation cephalosporin antibiotic, and its Spearman similarity score was 0.565. We examined whether these two drugs exhibited deleterious effects on TAAD in a BAPN-induced mouse model. C57BL/6J male mice aged 3 weeks were fed a normal laboratory diet and administered freshly prepared BAPN solution dissolved in the drinking water (0.5 g/kg per day) for consecutive 28 days. The mice were randomly divided into 7 groups: control, panobinostat, cefixime, BAPN, BAPN+ciprofloxacin, BAPN+panobinostat, and BAPN+cefixime. After 28 days, panobinostat and cefixime alone had no obvious changes compared with the control group. BAPN administration caused 33% (12/36) incidence of TAAD, including 11% (4/36) rupture and premature death in mice. Consistent with the previous report in which ciprofloxacin exacerbated TAAD in Ang II–infused mice,3 the application of ciprofloxacin in mice showed an increasing trend of the incidence of BAPN-induced TAAD (67% [8/12; P=0.088] incidence), including 17% (2/12; P=0.22) rupture and premature death in mice, and used it as a positive control. Intriguingly, the incidence of TAAD in mice of the BAPN+panobinostat group was increased to 76% (16/21; P=0.0025), including 29% (6/21; P=0.082) rupture and premature death, compared with the BAPN group. In contrast, cefixime (42% [5/12; P=0.73] incidence) did not aggravate BAPN-induced TAAD in mice and had no rupture and premature death (Figure [B and C]). Furthermore, the thoracic aortas from the mice of the BAPN+panobinostat group were more significantly expanded compared with the BAPN group (BAPN versus BAPN+panobinostat: ascending aortas, 1.47±0.48 versus 2.27±1.09 mm, P=0.0015; aortic arch, 1.54±0.34 versus 2.09±1.06 mm, P=0.041; descending aortas, 1.24±0.12 versus 1.75±0.80 mm, P=0.010; Figure [D]). Meanwhile, panobinostat aggravated elastin degradation and macrophage infiltration in aortic walls of TAAD induced by BAPN, as evidenced by elastic Van Gieson staining and immunofluorescent staining of CD68 (biomarker of macrophages), respectively (Figure [E and F]).Download figureDownload PowerPointFigure. pan-HDAC (histone deacetylase) inhibitors increase susceptibility of thoracic aortic aneurysm.A, The Connectivity Map (CMap) schematic diagram and the rational of drug screening from Connectivity Map database. The rainbow represents gene expression signature in response to the certain drug. Similar rainbow patterns represent the drugs have similar gene expression profiles. B–F, Three-week-old C57BL/6J male mice were administered vehicle (n=9), panobinostat (20 mg/kg 3 times per wk, n=12), TSA (trichostatin A; 0.6 mg/kg 3 times per wk, n=9), SAHA (vorinostat; 25 mg/kg daily for 14 d, n=9), cefixime (100 mg/kg per d, n=9); MC-1568 (50 mg/kg 3 times per wk, n=9), MS-275 (20 mg/kg 3 times per wk, n=9), BAPN (β-aminopropionitrile; 0.5 g/kg per d, n=36), BAPN+ciprofloxacin (100 mg/kg per d, n=12), BAPN+panobinostat (20 mg/kg 3 times per wk, n=21), BAPN+TSA (0.6 mg/kg 3 times per wk, n=14), BAPN+SAHA (25 mg/kg daily for 14 d, n=15), BAPN+cefixime (100 mg/kg per d, n=12), BAPN+MC-1568 (50 mg/kg 3 times per wk, n=15), or BAPN+MS-275 (20 mg/kg 3 times per wk, n=12) for 28 d. All mice were given both i.p. and i.g. administration simultaneously. For i.p. administration of panobinostat, TSA, SAHA, and MS-275, we also performed the vehicle i.g. administration. Conversely, the vehicle i.p. administration was performed in mice with i.g. administration of cefixime, ciprofloxacin, and MC-1568. The control- and BAPN-only groups were given both i.p. and i.g. administrations of vehicle. The body weight and the water intake of mice were measured every 2 d. The volume of water intake were ≈4.23 mL/d and 2.25 mL/d per mouse in control and BAPN group, respectively. The amount of water intake was comparable among all groups with BAPN administration, indicating BAPN was administered equally to mice. B, Representative ex vivo morphology of the aortas from the above 15 groups. C, Incidence of TAAD (thoracic aortic aneurysm and dissection; upper). Statistical analysis performed by Fisher exact test. Survival curve of mice with the indicated administration (lower). Statistical analysis performed by Log-rank test for Kaplan-Meier curve. D, Quantification of maximal aortic diameters measured ex vivo. Statistical analysis performed by 1-way ANOVA. E, Representative images of elastin Van Gieson staining of aortas from mice with the indicated administration (left) and quantification of elastin degradation grade (right, n=6). Nonparametric Kruskal-Wallis test with a Dunn multiple comparisons test was applied for comparison of the elastin degradation grade. Scale bar, 50 μm. The black arrows indicate the fragmented elastin. F, Representative images of immunofluorescence staining and quantification for CD68 (biomarker of macrophages)-positive (red) in aortas from mice with the indicated administration (n=10 for BAPN+TSA and n=6 for the other groups). Two-way ANOVA was applied to compare the macrophage infiltration. Nuclei were stained with DAPI (4',6-diamidino-2-phenylindole; blue). Scale bar, 25 μm. The white arrows indicate CD68-positive cells. Animal studies were approved by the Animal Care and Use Committee of Peking University. All data have been analyzed for normality and equal variance as a justification for using parametric analysis. Statistical analysis was performed using GraphPad Prism 8.0 software (GraphPad Software, San Diego, CA). Ad indicates adventitia; Arch, aortic arch; Asc, ascending; Des, descending; and Lu, lumen.To further examine effects of pan-HDAC inhibitors on TAAD progression, we explored another 2 pan-HDAC inhibitors vorinostat (SAHA) and trichostatin A (TSA) in BAPN-induced TAAD model. Consequently, both TSA and SAHA significantly increased the incidence of BAPN-induced TAAD (BAPN+TSA, 79% [11/14; P=0.0053]; BAPN+SAHA, 67% [10/15; P=0.036]) and subsequently exacerbated aortic elastin degradation and macrophage infiltration as well (Figure [B, C, E, and F]). Inhibition of the HDAC activity has been proposed as a promising therapeutic avenue in the treatment of cancers or cardiovascular diseases. Selective HDAC inhibitors such as class I selective HDAC inhibitors MCT-1 and MS-275 and class II selective HDAC inhibitor MC-1568 have been reported to reduce the pathogenesis and progression of abdominal aortic aneurysm in Ang II–infused ApoE−/− mice.5 However, our results identified that MS-275 and MC-1568 had no effect on BAPN-induced TAAD (Figure [B through D]). Beyond the HDAC family members targeted by MS-275 and MC-1568, pan-HDAC inhibitors additionally inhibit HDAC2, 8, 10, and 11. The inhibition of these HDACs might mediate the pathogenic effect of pan-HDAC inhibitors in TAAD, which requires further investigation.Our study pointed out that pan-HDAC inhibitors like panobinostat, TSA, and SAHA increased the susceptibility of TAAD in mice. Of note, panobinostat, TSA, and SAHA alone did not induce spontaneous TAAD in mice, but each of them increased the incidence and severity of BAPN-induced TAAD. In accordance, our finding urged that the patients with TAAD or individuals at risk for developing TAAD should be precautious with pan-HDAC inhibitor treatment in their medical therapies.Article InformationAcknowledgmentsQ. Cui and W. Kong designed the experiments. C. Huang curated the data set and performed the bioinformatics analysis with supervision from Q. Cui. S. Zhang and Z. Liu conducted the animal experiments and other chemical experiments with supervision from F. Yu and Y. Fu. Z. Liu helped in elastic Van Gieson staining. S. Zhang, Z. Liu, N. Xie, and W. Kong wrote the manuscript. All authors read and approved the final manuscript.Sources of FundingThis research was supported by funding from the National Natural Science Foundation of China (81730010, 91839302, 81921001, and 31930056) and the National Key R&D Program of China (2019YFA0801600).DisclosuresNone.Footnotes*S. Zhang, Z. Liu, and N. Xie contributed equally.For Sources of Funding and Disclosures, see page 2850.Correspondence to: Wei Kong, MD, PhD, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China, Email [email protected]edu.cnQinghua Cui, PhD, Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China, Email [email protected]pku.edu.cnReferences1. Daneman N, Lu H, Redelmeier DA. Fluoroquinolones and collagen associated severe adverse events: a longitudinal cohort study.BMJ Open. 2015; 5:e010077. doi: 10.1136/bmjopen-2015-010077CrossrefMedlineGoogle Scholar2. Lee CC, Lee MT, Chen YS, Lee SH, Chen YS, Chen SC, Chang SC. Risk of aortic dissection and aortic aneurysm in patients taking oral fluoroquinolone.JAMA Intern Med. 2015; 175:1839–1847. doi: 10.1001/jamainternmed.2015.5389CrossrefMedlineGoogle Scholar3. LeMaire SA, Zhang L, Luo W, Ren P, Azares AR, Wang Y, Zhang C, Coselli JS, Shen YH. Effect of ciprofloxacin on susceptibility to aortic dissection and rupture in mice.JAMA Surg. 2018; 153:e181804. doi: 10.1001/jamasurg.2018.1804CrossrefMedlineGoogle Scholar4. Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, Lerner J, Brunet JP, Subramanian A, Ross KN, et al.. The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease.Science. 2006; 313:1929–1935. doi: 10.1126/science.1132939CrossrefMedlineGoogle Scholar5. Galán M, Varona S, Orriols M, Rodríguez JA, Aguiló S, Dilmé J, Camacho M, Martínez-González J, Rodriguez C. Induction of histone deacetylases (HDACs) in human abdominal aortic aneurysm: therapeutic potential of HDAC inhibitors.Dis Model Mech. 2016; 9:541–552. doi: 10.1242/dmm.024513CrossrefMedlineGoogle Scholar Previous Back to top Next FiguresReferencesRelatedDetailsCited By Ran J, Zhang Y, Zhang S, Li H, Zhang L, Li Q, Qin J, Li D, Sun L, Xie S, Zhang X, Liu L, Liu M and Zhou J (2022) Targeting the HDAC6‐Cilium Axis Ameliorates the Pathological Changes Associated with Retinopathy of Prematurity, Advanced Science, 10.1002/advs.202105365, (2105365) Qian G, Adeyanju O, Olajuyin A and Guo X (2022) Abdominal Aortic Aneurysm Formation with a Focus on Vascular Smooth Muscle Cells, Life, 10.3390/life12020191, 12:2, (191) November 2021Vol 41, Issue 11Article InformationMetrics © 2021 American Heart Association, Inc.https://doi.org/10.1161/ATVBAHA.121.316808PMID: 34525846 Originally publishedSeptember 16, 2021 Keywordsaortic dissectionhistone deacetylase inhibitorsthoracic aortic aneurysmPDF download Advertisement SubjectsAneurysmAortic DissectionTranslational StudiesVascular Biology
What problem does this paper attempt to address?
-
Abstract 17364: Lysyl Oxidase Inhibition Induces Acute Aortic Dissection Through Activation of Focal Adhesions and Mammalian Target of Rapamycin Signaling in Mice
Zhen Zhou,Pujun Guan,Ripon Sarkar,Zhouxuan Li,Dongchuan Guo,Xue Yan Duan,Callie S Kwartler,Dianna M Milewicz
DOI: https://doi.org/10.1161/circ.148.suppl_1.17364
IF: 37.8
2023-11-07
Circulation
Abstract:Introduction: Defective genes that predispose to acute aortic dissections (AADs) suggest that maintenance of aortic integrity requires mature extracellular matrix (ECM) and intracellular homeostatic contraction of smooth muscle cells (SMCs). Focal adhesions (FAs) link the ECM to SMC contractile units, and we sought to determine the role of SMC FA signaling in a mouse model in which AADs are induced by disrupting ECM with a lysyl oxidase inhibitor, β-aminopropionitrile (BAPN). Methods: C57BL/6J wild-type mice at postnatal day 21 (P21) were exposed to BAPN to induce AADs. All aortas were visually inspected at necropsy or tissue harvest. Proximal aortas were harvested for single-cell RNA sequencing (scRNAseq) at P35 and for immunoblot analyses at P24 and P35. Echocardiography, tail-cuff blood pressure, and histopathology analyses were performed on surviving mice at P49. Results: MELD analysis of scRNAseq data identified SMC clusters as the top altered cell type in BAPN-treated aortas. KEGG pathway analysis identified activated FA, along with Rho/ROCK and mTOR signaling, in the BAPN-treated SMC cluster. Immunoblots confirmed increased phosphorylation of FAK, MYPT1, mTOR, p70S6K, and S6RP in BAPN-treated aortas, and FAK inhibition blocked Rho/ROCK and mTOR pathways. Pharmacological inhibition of mTOR with rapamycin significantly rescued BAPN-induced AAD deaths (9% vs. 50%, p=0.001) without altering blood pressure, aortic enlargement, or aortic medial elastic fiber degeneration. Immunoblot analyses confirmed dramatic decrease of mTOR, p70S6K, and S6RP phosphorylation levels in BAPN aortas with rapamycin treatment. Administration of a ROCK inhibitor, Y-27632, significantly augmented type A AAD incidence (p=0.04) without altering blood pressure in BAPN mice, and immunoblot analyses identified increased mTOR signaling as early as P24 in BAPN plus Y-27632-treated aortas compare with BAPN group. Conclusions: Activation of FA and downstream mTOR signaling triggers AAD when the ECM is disrupted by BAPN, and mTOR inhibition significantly attenuates these deaths. FAs also activate Rho/ROCK, which is protective. These findings support that preventing mTOR signaling or augmenting Rho/ROCK signaling are therapeutic targets to prevent AADs.
cardiac & cardiovascular systems,peripheral vascular disease
-
Abstract 277: The Effect Of Phosphodiesterase Inhibitors On Bapn Induced Aortic Dissection And Aneurysm
Sam Tyagi,Jacob Hubbuch,Michael Franklin,Deborah A Howatt,Hisashi Sawada,Hong S Lu,Alan Daugherty
DOI: https://doi.org/10.1161/atvb.43.suppl_1.277
2023-05-01
Abstract:Introduction: Aortic dissection (AoD) is manifested by bleeding into the medial layers of the aortic wall. Aortic aneurysm (AA) is a dilation of the aorta. One fatal consequence of AoD and AA is rupture of the aortic wall. There are no pharmacological or therapeutic agents to decrease the risk of AA or to promote positive remodeling of the aorta after AoD. Phosphodiesterase inhibitors (PDEi) have diverse effects on cardiovascular diseases. There is experimental evidence that cilostazol (PDE3i) protects against AA, and alternatively there are clinical reports of sildenafil (PDE5i) inducing AoD. However, the role in PDEi’s in aortic pathology is still unclear. Our hypothesis is that sildenafil will exacerbate AoD, and cilostazol protects against AoD and AA. Our aim was to determine the effects of PDE3i and PED5i in β-aminoproprionitrile (BAPN) AoD and angiotensin II- (AngII) induced AAA models, respectively. Methods and Results: BAPN was administered to induce AoD formation in male C57BL/6J mice. We analyzed ascending and descending aortic tissue to determine if there was increased expression of known PDE related genes using RNA sequencing in mice administered either vehicle or BAPN for 7 days mice. We found increased PDE3 mRNA abundance in the ascending aorta by 19% (P=0.006), and increased of PDE5 mRNA abundance in the descending aorta by 21% (P=0.048). Subsequently, BAPN was administered in drinking water in 4-week-old male C57BL/6J mice for 12 weeks. There were three groups of mice: regular diet, and diets containing cilostazol or sildenafil. There had no difference on death rate due to aortic rupture between the groups. After termination, the ascending, descending and abdominal aortas were measured in situ, and there was no difference in aortic diameters. In a separate study, AngII osmotic pumps were inserted in male ApoE-/- mice to induced aortic aneurysm using the three there was no difference in the mortality rates. After 4 weeks of AngII infusion, the aorta was measured in situ, and there was no difference in aortic diameters. Conclusion: Inhibition of either PDE3 or PDE5 has no effects on either BAPN-induced AoD or AngII-induced AAA.
peripheral vascular disease,hematology
-
β-Aminopropionitrile Induces Distinct Pathologies in the Ascending and Descending Thoracic Aortic Regions of Mice
Michael K. Franklin,Hisashi Sawada,Sohei Ito,Deborah A. Howatt,Naofumi Amioka,Ching-Ling Liang,Nancy Zhang,David B. Graf,Jessica J. Moorleghen,Yuriko Katsumata,Hong S. Lu,Alan Daugherty
DOI: https://doi.org/10.1161/atvbaha.123.320402
2024-05-25
Arteriosclerosis Thrombosis and Vascular Biology
Abstract:Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. BACKGROUND:β-aminopropionitrile (BAPN) is a pharmacological inhibitor of LOX (lysyl oxidase) and LOXLs (LOX-like proteins). Administration of BAPN promotes aortopathies, although there is a paucity of data on experimental conditions to generate pathology. The objective of this study was to define experimental parameters and determine whether equivalent or variable aortopathies were generated throughout the aortic tree during BAPN administration in mice.METHODS:BAPN was administered in drinking water for a period ranging from 1 to 12 weeks. The impacts of BAPN were first assessed with regard to dose, strain, age, and sex. BAPN-induced aortic pathological characterization was conducted using histology and immunostaining. To investigate the mechanistic basis of regional heterogeneity, the ascending and descending thoracic aortas were harvested after 1 week of BAPN administration before the appearance of overt pathology.RESULTS:BAPN-induced aortic rupture predominantly occurred or originated in the descending thoracic aorta in young C57BL/6J or N mice. No apparent differences were found between male and female mice. For mice surviving 12 weeks of BAPN administration, profound dilatation was consistently observed in the ascending region, while there were more heterogeneous changes in the descending thoracic region. Pathological features were distinct between the ascending and descending thoracic regions. Aortic pathology in the ascending region was characterized by luminal dilatation and elastic fiber disruption throughout the media. The descending thoracic region frequently had dissections with false lumen formation, collagen deposition, and remodeling of the wall surrounding the false lumen. Cells surrounding the false lumen were predominantly positive for α-SMA (α-smooth muscle actin). One week of BAPN administration compromised contractile properties in both regions equivalently, and RNA sequencing did not show obvious differences between the 2 aortic regions in smooth muscle cell markers, cell proliferation markers, and extracellular components.CONCLUSIONS:BAPN-induced pathologies show distinct, heterogeneous features within and between ascending and descending aortic regions in mice.
peripheral vascular disease,hematology
-
Abstract 268: Exercise Modulates Inflammatory, Cytokine, And Growth Factor Pathway Dysregulation In A Mouse Model Of Aortic Dissection And Aneurysm
Matthew D Cahn,Jackie M Zhang,Fenge Ni,Brajesh Lal,Dudley Strickland,Areck Ucuzian
DOI: https://doi.org/10.1161/atvb.43.suppl_1.268
2023-05-01
Abstract:Aerobic exercise can prevent aortopathy in mouse models. The objective of this study was to better understand the mechanisms underlying this finding. Mice treated with β-aminopropionitrile (BAPN), an irreversible inhibitor of lysyl oxidase (LOX) which induces thoracic aortic aneurysm and dissection (TAAD), underwent either a forced moderate treadmill regimen or maintained conventional cage activity without forced exercise. RNA sequencing was utilized to determine the transcriptomic profile of the ascending and thoracic aorta (n=7 per group). BAPN induced overexpression of 357 genes and decreased expression of 77 genes compared with controls. Causal analysis in Ingenuity Pathway Analysis® software using a fold change |2| and FDR 2, p-value 2, BH p-value of overlap < 0.05). A comparison analysis between the two groups sorted by trend and score demonstrates that moderate aerobic exercise reverses the predicted activation of these pathways. Next, we investigated whether activated TGFβ1 signaling, predicted to be normalized with exercise compared with unexercised mice, is required for the aortopathy caused by LOX inhibition. We administered SB431542, a selective small peptide inhibitor of the TGF-β1 receptor (TGF-β1R), to a separate cohort of BAPN-treated mice. Histologic analysis demonstrated that the addition of the TGF-β1R inhibitor reverses many of the histologic changes characteristic of BAPN-induced aortopathy, including reversal of wall thickening (p < 0.001), elastin degradation (p < 0.01), and cystic medial degeneration (p = 0.04) in the thoracic aorta. While further investigation is necessary, our study suggests that the prevention of TAAD and its complications by moderate aerobic exercise may in part be due to deactivation of the dysregulated TGF-β1 pathway.
peripheral vascular disease,hematology
-
Oltipraz, the activator of nuclear factor erythroid 2-related factor 2 (Nrf2), protects against the formation of BAPN-induced aneurysms and dissection of the thoracic aorta in mice by inhibiting activation of the ROS-mediated NLRP3 inflammasome
Dashuai Wang,Jia Wu,Sheng Le,Hongfei Wang,Jingjing Luo,Rui Li,Xing Chen,Long Wu,Ping Ye,Xinling Du,Xiaofan Huang,Song Yu
DOI: https://doi.org/10.1016/j.ejphar.2022.175361
IF: 5.195
2022-11-05
European Journal of Pharmacology
Abstract:Background Thoracic aortic aneurysm and dissection (TAAD) is caused by the apoptosis and phenotypic transformation of vascular smooth muscle cells (VSMCs). The dysfunction of VSMCs affects their secretion of chemokines such as monocyte chemoattractant protein-1 (MCP-1) to recruit the infiltration of macrophages which release proinflammatory cytokines and matrix metalloproteinases (MMPs) to accelerate the process of TAAD formation. Approach and results We analyzed the expression levels of nuclear factor erythroid 2-related factor 2 (Nrf2) in aortic tissues of TAAD patients and the β-aminopropionitrile fumarate (BAPN)-induced mouse model, and the levels of Nrf2 were elevated in both aortic lesions. Treatment with the Nrf2 activator oltipraz protects against the formation of BAPN-induced aneurysm and dissection, as demonstrated by a higher survival rate, postponing the time of aortic rupture, and inhibiting aortic luminal dilation. In addition, the thoracic aortas of BAPN-treated mice inhibited the apoptosis and phenotypic transformation of VSMCs. When treated with oltipraz, they had reduced macrophage infiltration proinflammatory cytokines and MMPs. Furthermore, oltipraz treatment promoted the translocation of Nrf2 and downregulated the NLRP3 pathway. Conclusion Nrf2 plays a crucial role in protecting against TAAD development, and persistent activation of Nrf2 is a promising therapeutic strategy against the progression of TAAD.
pharmacology & pharmacy
-
Abstract 543: Inhibition Of Renin-angiotensin System Failed To Suppress Beta-aminopropionitrile-induced Thoracic Aortopathies In Mice
Hisashi Sawada,Satoko Ohno,Dien Ye,Michael Franklin,Deborah A Howatt,Jessica J Moorleghen,Hong Lu,Alan Daugherty
DOI: https://doi.org/10.1161/atvb.42.suppl_1.543
2022-01-01
Abstract:Objective: Cross-linking of elastin and collagen fibers is a vital process in aortic development. Pharmacological inhibition of aortic cross-linking by β-aminopropionitrile (BAPN) leads to thoracic aortopathies, such as thoracic aortic aneurysm and dissection, in mice. Although the renin angiotensin system (RAS) contributes to several types of thoracic aortopathies, it remains unclear whether inhibition of the RAS protects against aortopathy formation caused by the impairment of elastin and collagen cross-linking in mice. Approach and Results: Either vehicle or losartan (15 mg/kg/d, AT1 receptor blocker) were administered through osmotic pumps for 4 weeks in male C57BL/6J mice (3-4-weeks-old, n=30/group). BAPN (0.5% wt/vol) was given through drinking water to induce aortopathies. Losartan increased plasma renin concentrations significantly compared to vehicle-infused mice, indicating effective blockade of at1 receptors. However, losartan did not suppress BAPN-induced aortic rupture and dilatation. Since losartan is a surmountable inhibitor, we also tested the effects of irbesartan, an insurmountable inhibitor of AT1 receptor blocker, on BAPN-induced aortopathy formation (50 mg/kg/day, diet, n=30/group). Despite the significant increase of plasma renin concentrations, irbesartan did not ameliorate aortic rupture and dilatation in BAPN-administered mice. Thus, BAPN-induced thoracic aortopathies were refractory to angiotensin receptor blockade. We next inhibited angiotensinogen, the unique substrate for angiotensin II, using an antisense oligonucleotide targeting angiotensinogen (AGT-ASO). AGT-ASO decreased plasma angiotensinogen concentrations, while aortic death and dilatations were not attenuated. Since hepatocytes are the major source of angiotensinogen, BAPN-induced thoracic aortopathy formation was also examined in mice with hepatocyte-specific angiotensinogen deletion. Although plasma angiotensinogen concentrations displayed 90% reduction compared to wild type littermates, aortic rupture and aneurysm formation were not suppressed. Conclusion: Inhibition of the RAS does not attenuate BAPN-induced thoracic aortopathy formation in mice.
-
Effect of Ciprofloxacin on Susceptibility to Aortic Dissection and Rupture in Mice
Scott A. LeMaire,Lin Zhang,Wei Luo,Pingping Ren,Alon R. Azares,Yidan Wang,Chen Zhang,Joseph S. Coselli,Ying H. Shen
DOI: https://doi.org/10.1001/jamasurg.2018.1804
2018-09-19
JAMA Surgery
Abstract:Importance: Fluoroquinolones are among the most commonly prescribed antibiotics. Recent clinical studies indicated an association between fluoroquinolone use and increased risk of aortic aneurysm and dissection (AAD). This alarming association has raised concern, especially in patients with AAD with risk of rupture and in individuals at risk for developing AAD.Objective: To examine the effect of ciprofloxacin on AAD development in mice.Design, Setting, and Participants: In a mouse model of moderate, sporadic AAD, 4-week-old male and female C57BL/6J mice were challenged with a high-fat diet and low-dose angiotensin infusion (1000 ng/min/kg). Control unchallenged mice were fed a normal diet and infused with saline. After randomization, challenged and unchallenged mice received ciprofloxacin (100 mg/kg/d) or vehicle through daily gavage during angiotensin or saline infusion. Aortic aneurysm and dissection development and aortic destruction were compared between mice. The direct effects of ciprofloxacin on aortic smooth muscle cells were examined in cultured cells.Results: No notable aortic destruction was observed in unchallenged mice that received ciprofloxacin alone. Aortic challenge induced moderate aortic destruction with development of AAD in 17 of 38 mice (45%) and severe AAD in 9 (24%) but no rupture or death. However, challenged mice that received ciprofloxacin had severe aortic destruction and a significantly increased incidence of AAD (38 of 48 [79%]; P = .001; χ2 = 10.9), severe AAD (32 of 48 [67%]; P < .001; χ2 = 15.7), and rupture and premature death (7 of 48 [15%]; P = .01; χ2 = 6.0). The increased AAD incidence was observed in different aortic segments and was similar between male and female mice. Compared with aortic tissues from challenged control mice, those from challenged mice that received ciprofloxacin showed decreased expression of lysyl oxidase, an enzyme that is critical in the assembly and stabilization of elastic fibers and collagen. These aortas also showed increased matrix metalloproteinase levels and activity, elastic fiber fragmentation, and aortic cell injury. In cultured smooth muscle cells, ciprofloxacin treatment significantly reduced lysyl oxidase expression and activity, increased matrix metalloproteinase expression and activity, suppressed cell proliferation, and induced cell death. Furthermore, ciprofloxacin-a DNA topoisomerase inhibitor-caused nuclear and mitochondrial DNA damage and the release of DNA into the cytosol, subsequently inducing mitochondrial dysfunction, reactive oxygen species production, and activation of the cytosolic DNA sensor STING, which we further showed was involved in the suppression of lysyl oxidase expression and induction of matrix metalloproteinase expression.Conclusions and Relevance: Ciprofloxacin increases susceptibility to aortic dissection and rupture in a mouse model of moderate, sporadic AAD. Ciprofloxacin should be used with caution in patients with aortic dilatation, as well as in those at high risk for AAD.
surgery
-
Involvement of B cells in the pathophysiology of β-aminopropionitrile-induced thoracic aortic dissection in mice
Yanxiang Gao,Zhizhi Wang,Jianqiao Zhao,Weiliang Sun,Jing Guo,Zufang Yang,Yimin Tu,Changan Yu,Lin Pan,Jingang Zheng
DOI: https://doi.org/10.1538/expanim.18-0170
IF: 2.4
2019-01-01
Experimental Animals
Abstract:Thoracic aortic dissection (TAD) is a life-threatening disease that is characterized by an inflammatory response. Innate and cellular immunity has long been known to be involved in TAD, but the role of humoral immunity in the pathophysiology of TAD remains unknown. We administered the lysyl oxidase inhibitor β-aminopropionitrile (BAPN; 1 g/kg/day) in 3-week-old male C57BL/6J mice for 4 weeks to establish an animal model of TAD. Animals that died were immediately dissected. Animals that survived were sacrificed on days 7, 14, and 28 after BAPN challenge. The incidence and rupture rates of BAPN-induced TAD were 90% (9/10) and 70% (7/10), respectively, at 28 days. Victoria blue-nuclear fast red staining of aortic tissue revealed elastic lamellae destruction and the formation of a false lumen in the BAPN group. Hematoxylin-eosin staining revealed the infiltration of both plasmacytoid mononuclear cells and polymorphonuclear inflammatory cells in TAD tissues. Enzyme-linked immunosorbent assay and immunohistochemistry indicated that plasma immunoglobin M (IgM) and IgG were elevated at 7, 14, and 28 days, and CD19-positive B cells infiltrated into the adventitia of aortic tissue in BAPN-treated mice. The transcriptional analysis showed an increase in the expression of B cell receptor signaling-associated genes. These results indicate that B cells and immunoglobulins might participate in the pathogenesis of TAD, suggesting that humoral immunity may be a possible therapeutic target for TAD.
zoology,veterinary sciences
-
Urate-Lowering Therapy Inhibits Thoracic Aortic Aneurysm and Dissection Formation in Mice
Liu Yang,Hao Wu,Congcong Luo,Yang Zhao,Rongbo Dai,Zhiqing Li,Xu Zhang,Ze Gong,Zeyu Cai,Yicong Shen,Fang Yu,Wei Li,Hongmei Zhao,Tao Zhang,Junming Zhu,Yi Fu,Jing Wang,Wei Kong
DOI: https://doi.org/10.1161/ATVBAHA.122.318788
2023-04-29
Abstract:Arteriosclerosis, Thrombosis, and Vascular Biology, Ahead of Print. Background:Thoracic aortic aneurysm and dissection (TAAD) is a highly lethal vascular disease without effective drug therapy. Whether elevated serum concentrations of uric acid are involved in TAAD development remains unclear.Methods:Serum uric acid levels were detected in different TAAD mouse models and patients. The urate-lowering drug allopurinol was administered in the drinking water of TAAD mice. Adenine diet–induced mice were established to investigate the role of hyperuricemia in TAAD formation and RNA-sequencing of thoracic aortas from these mice was performed.Results:We found serum uric acid levels were elevated in various mouse TAAD models, including mice fed a β-aminopropionitrile diet, Marfan mice with fibrillin-1 haploinsufficiency (Fbn1C1041G/+), and ApoE−/−mice infused with Ang II (angiotensin II), as well as in patients with TAAD. Administration of urate-lowering drug allopurinol in the drinking water significantly alleviated TAAD formation in β-aminopropionitrile–treated mice, Fbn1C1041G/+mice, and Ang II–infused ApoE−/−mice. Moreover, an adenine diet was used to induce hyperuricemia in mice. Intriguingly, a 4-week adenine diet feeding directly induced TAAD formation characterized by increased maximal thoracic aortic diameters and severe elastin degradation, which were ameliorated by allopurinol. Unbiased RNA-sequencing in mouse thoracic aortas suggested that FcγR (Fc gamma receptor) was upregulated upon adenine diet, but reciprocally repressed by allopurinol. Mechanistically, hyperuricemia activated FcγR-mediated ERK1/2 (extracellular signal-regulated kinase 1/2) phosphorylation to induce macrophage inflammation and TAAD development, which was abrogated by allopurinol or FcγR deficiency.Conclusions:This study uncovered an important and previously unrecognized role of hyperuricemia in mediating the pathogenesis of TAAD, and uric acid–lowering drug may represent a promising therapeutic approach for TAAD.
peripheral vascular disease,hematology
-
Abstract 1012: Single-Cell RNA Sequencing Defines Focal Adhesion-Matrix Interactions In Smooth Muscle Cells In Thoracic Aortic Disease Triggered By BAPN Inhibiting Lysyl Oxidase
Pujun Guan,Zhen Zhou,Ripon Sarkar,Dongchuan Guo,Xue Yan Duan,Callie Kwartler,Dianna M Milewicz
DOI: https://doi.org/10.1161/atvb.44.suppl_1.1012
2024-05-01
Abstract:Background: Thoracic aortic aneurysms and/or dissections (TAD) are associated with pathogenic variants in genes that involved in either smooth muscle cell (SMC) contraction or extracellular matrix (ECM). The inability to maintain homeostasis between SMCs and ECM is pivotal in TAD pathogenesis. Focal adhesions (FA) act as mechanosensors in cells, transferring mechanical signals from the ECM to the intracellular contractile filaments and regulating downstream Rho/ROCK signaling. However, the landscape of SMC-ECM interactions, has not been fully explored at the early stages of TAD. Methods: C57BL/6J wild-type mice aged 21 days postnatal (P21) were administered the ECM cross-link blocker, BAPN, for up to four weeks to induce TAD. Proximal aortas were harvested for single-cell RNA sequencing at P35. Immunofluorescence and immunoblots were used to validate findings. Results: Single-cell RNA sequencing classified SMCs into 5 clusters marked by Rgs5 (SMC Rgs5 + ), high levels of Palld (SMC Palld + ), Tnnt2 (SMC Tnnt2 + , marker of second heart field-derived cells), Atf3 (SMC Atf3+ cluster), or no clear marker (SMC NOS ). The analysis of how BAPN exposure affects different cell types, based on the Jensen-Shannon divergence in the probability distribution between groups, demonstrated that SMCs, and specifically SMC Atf3 + and SMC NOS , are the most perturbed cell cluster by BAPN. Gene set enrichment analysis identified significantly dysregulated pathways of FA and Rho/ROCK signaling. The expression of Itga5 and Fn1 (fibronectin) were increased and Itga8 was decreased. An SMC-SMC interaction analysis found Fn1 interactions were the most upregulated interaction in 4 SMC clusters. When analyzing downstream pathways due to the changes in interaction, 80% of the top 25 interactions were related to integrins, including increased Fn1 interactions with Itga5-Itgb1 and Itgav-Itgb3. In SMC Rgs5 + cluster, the most affected interactions are related to TGF-β signaling. Conclusions: These findings suggest that, in the majority of aortic SMCs, mechanosensing through FAs, driven by fibronectin interactions with specific integrin receptor pairs, is activated with BAPN treatment, potentially contributing to the pathogenesis of TAD.
peripheral vascular disease,hematology
-
BAPN-induced rodent model of aortic dissecting aneurysm and related complications
Tongyun Chen,Nan Jiang,Shaopeng Zhang,Qingliang Chen,Zhigang Guo
DOI: https://doi.org/10.21037/jtd-21-605
2021-06-01
Journal of Thoracic Disease
Abstract:BACKGROUND: The aim of this study was to investigate the effects of beta-aminopropionitrile (BAPN) on the arterial walls of rodents, and to analyze the gross or pathological changes of arterial and other tissues of rodents treated with BAPN at different concentrations or doses.METHODS: Eighteen SPF SD rats (4-5-week old) were divided into three groups: SD-0.2 (Group A), SD-0.4 (Group B), and SD-0.6 (Group C). The groups A, B and C were given 0.2%, 0.4%, and 0.6% BAPN solution, respectively, as drinking water for seven weeks. Forty SPF C57BL/6 mice (3-week old) were randomly divided into four groups: C57-0.2 (Group D), C57-0.4 (Group E), C57-0.6 (Group F) and the control group and given 0.2%, 0.4%, or 0.6% BAPN or distilled water as drinking water, respectively, for seven weeks. All experimental animals were free to drink water. The aortas were dissected and visually examined. At the same time, hematoxylin and eosin (HE) staining was performed in aorta tissue. The vascular diameter and area of the middle membrane were measured with IPP (Image-Pro Plus 6.0).RESULTS: BAPN treatment significantly affected the water intake and weight gain of rats and mice. BAPN also caused thickening of the membrane in the aortas of rats and mice, and irregularity in the arrangement of elastic fibers. These pathological changes are similar to the pathological changes observed in human aneurysms. The incidence of dissecting aneurysm in C57 mice was higher than that of Sprague Dawley (SD) rats.CONCLUSIONS: BAPN at a concentration of 0.4% was feasible to produce an animal model of dissecting aneurysm. In SD rats, the rate of pathological changes and other complications, such as intestinal rupture and scoliosis, was higher than the rates of dissecting aneurysm.
respiratory system
-
Postnatal Deficiency of ADAMTS1 Ameliorates Thoracic Aortic Aneurysm and Dissection in Mice
Shanshan Wang,Yuting Liu,Guizhen Zhao,Li He,Yi Fu,Changan Yu,Zhizhi Wang,Tingting Zhao,Fan Cao,Yanxiang Gao,Wei Kong,Jingang Zheng
DOI: https://doi.org/10.1113/ep087018
2018-01-01
Experimental Physiology
Abstract:What is the central question of this study?Thoracic aortic aneurysm and dissection (TAAD) is characterized by extracellular matrix remodelling and an inflammatory response. Evidence suggests that ADAMTS1 is closely associated with TAAD development, but whether it contributes to the pathophysiology of TAAD remains unknown. What is the main finding and its importance?We generated inducible postnatal ADAMTS1 knockout mice and found that ADAMTS1 deficiency attenuated beta-aminopropionitrile-dependent TAAD formation and rupture. Furthermore, ADAMTS1 deficiency suppressed neutrophil and macrophage infiltration by inhibiting inflammatory cytokine levels and macrophage migration during the early stage of beta-aminopropionitrile-induced TAAD. ADAMTS1 could be a new therapeutic target for TAAD. Thoracic aortic aneurysm and dissection (TAAD), as a life-threatening cardiovascular disease, is characterized by extracellular matrix remodelling and an inflammatory response. A disintegrin and metalloproteinase with thrombospondin motifs 1 (ADAMTS1) is an inflammation-related protein that is able to degrade extracellular matrix proteins in arteries. Herein, we investigated whether ADAMTS1 contributes to the pathophysiology of TAAD in mice. Using the mouse model of beta-aminopropionitrile (BAPN)-induced TAAD, we found that ADAMTS1 expression was upregulated beginning in the early stage of TAAD development and localized predominantly in the aortic adventitia. ADAMTS1-floxed mice and whole-body tamoxifen-inducible ADAMTS1 knockout mice (ADAMTS1(flox/flox) Ubc-Cre(ERT2+), ADAMTS1 KO) were generated to investigate the direct causal role of ADAMTS1 in TAAD development. The incidence and rupture rates of BAPN-induced TAAD in ADAMTS1 KO mice were significantly lower than those in ADAMTS1(flox/flox) mice (45.5 versus 81.8% and 18.2 versus 42.4%, respectively). Aortas from BAPN-treated ADAMTS1(flox/flox) mice displayed profound destruction of the elastic lamellae, abundant neutrophil and macrophage accumulation in the adventitia, obviously increased neutrophil proportions in peripheral blood and significantly increased expression of inflammatory factors in the early stage of TAAD induction, all of which were markedly suppressed in ADAMTS1 KO mice. Furthermore, ADAMTS1-deficient macrophages exhibited abrogated migration capacity both in vivo and in vitro. In conclusion, ADAMTS1 plays a crucial role in postnatal TAAD formation and rupture by regulating inflammatory responses, suggesting that ADAMTS1 might be a new therapeutic target for TAAD.
-
PANoptosis is a prominent cell death feature in thoracic aortic aneurysm or dissection
Xu Xu,Yaxin Zhu,Yuting Niu,Yufei Chen,Siyang Fan,Dingkun Lu,Ruixia Xu,Xiaohan Fan
DOI: https://doi.org/10.1016/j.yexcr.2024.114247
2024-10-01
Abstract:Thoracic aortic aneurysm and dissection (TAAD) is a devastating macrovascular disease, and its pathogenic mechanisms have not been well clarified. This study aimed to investigate the role of PANoptosis, which is newly defined programmed cell death (PCD) and characterized by pyroptosis, apoptosis, and necroptosis, in the pathogenesis of TAAD. We found that the expression of initiator factor Z-DNA binding protein 1 (ZBP1) and PANoptosis-related genes were upregulated in the β-aminopropionitrile (BAPN) + Angiotensin II (Ang II)-induced TAAD mice. Ang II stimuli enhanced the expression of ZBP1, promoted the generation of bioactive GSDMD (Gasdermin D) fragments, the cleavage of Caspase 3, and increased the phosphorylation of mixed lineage kinase domain-like pseudokinase (MLKL) in human aortic vascular smooth muscle cells (HASMCs), indicating the activation of hallmarks for PANoptosis. Moreover, ZBP1-mediated PANoptosis occurs in the aortic tissues of TAAD patients. These results highlight the significant role of PANoptosis in TAAD pathogenesis, suggesting ZBP1 and other PANoptosis-related genes as potential therapeutic targets for this condition.
-
Abstract 316: Chronic Intermittent Hypoxia Attenuates BAPN Induced Thoracic Aortic Aneurysms in Mice
Yunyun Yang,Linyi Li,Yan Wen Qin
DOI: https://doi.org/10.1161/atvb.38.suppl_1.316
2018-05-01
Abstract:Objective: Aortic aneurysm (AA) is a life-threatening cardiovascular disorder due to the predisposition for dissection and rupture. Chronic intermittent hypoxia (CIH) has protective effects on heart and brain against ischemia injury. However, whether CIH prevents against AA formation was not elucidated. The present study aimed to investigate the effect of CIH treatment on thoracic aortic aneurysms (TAA) in mice. Approach and Results: Seventy-five C57BL/6 mice 3 weeks of age were divided into three groups, i.e. Control group ( 21% O 2 , 24 h per day, 4 weeks, n = 15); β-aminopropionitrile (BAPN) group (oral administration BAPN 1g / kg / day, 21% O 2 , 24 h per day, 4 weeks, n = 30); CIH+BAPN group ( 5% -21% O 2 90s / cycle, 8h per day, 4 weeks; BAPN 1g / kg / day, n = 30). We found that CIH significantly reduced the incidence of TAA (18 of 30 versus 9 of 30; p<0.05), and decreased maximal aortic diameter in BAPN group by hematoxylin and eosin staining (1.71 ± 0.35 versus 1.23± 0.15 mm; p <0.05). CIH significantly reduced elastin breaks in BAPN group by Verhoeff-van Gieson staining. The apoptosis of vascular smooth muscle cells was detected by TUNEL staining increased in BAPN group and markedly reversed by CIH. Moreover, RNA-Seq analysis and bioinformatics analysis were performed to screen the differential expression genes and possible pathway. CIH significantly down-regulated the expression of inflammation signaling pathways, up-regulated the expression of PI3K/AKT survival pathway and extracellular matrix homeostasis related pathway in BAPN induced TAA mice. Confirmed by real-time quantitative reverse-transcription-polymerase chain reaction (qRT-PCR) assays, CIH significantly inhibited the expression of IL-1β, TNF-α, MMP-9 and Cathepsin S, and increased the expression of hypoxia inducible factor-1α (HIF-1α), Lysyl oxidase (LOX), ITGB1 and COL1A2. Our findings also showed that the expression of MMP-9, MMP-2, cathepsin S, IL-1β, MCP-1 and Mac-2 increased in BAPN group and markedly decreased in CIH+BAPN group by immunohistochemistry staining. Besides, CIH also significantly increased the protein expression of HIF-1α, LOX and p-AKT by western blot analysis. Conclusions: The findings from our study suggest that CIH prevented the TAA development of BAPN-induced mice.
peripheral vascular disease,hematology
-
Rapamycin prevents thoracic aortic aneurysm and dissection in mice
Biao Zhou,Wei Li,Guizhen Zhao,Bing Yu,Baihui Ma,Zhujiang Liu,Nan Xie,Yi Fu,Ze Gong,Rongbo Dai,Xiaoming Zhang,Wei Kong
DOI: https://doi.org/10.1016/j.jvs.2018.05.246
Abstract:Objective: The purpose of this study was to investigate whether rapamycin inhibits the development of thoracic aortic aneurysm and dissection (TAAD) in mice. Methods: Three-week-old C57BL/6J male mice were fed a normal diet and randomized into a control group (n = 6), β-aminopropionitrile fumarate (BAPN) group (Gp A; n = 15), BAPN plus rapamycin (5 mg) group (Gp B; n = 8), and BAPN plus rapamycin (10 mg) group (Gp C; n = 8). Gp A, Gp B, and Gp C were administered BAPN (1 g/kg/d) for 4 weeks. One week after BAPN administration, Gp B and Gp C were treated with rapamycin (5 mg/kg/d or 10 mg/kg/d) through gavage for 21 days. Thoracic aortas were harvested for Western blot and immunofluorescence staining at day 14 and for morphologic and histologic analyses at day 28. Results: BAPN treatment induced TAAD formation in mice. The incidence of TAAD in control, Gp A, Gp B, and Gp C mice was 0%, 80%, 25%, and 37.5%, respectively. Smaller thoracic aortic diameters (ascending aorta and arch) were observed in Gp B and Gp C mice than in Gp A mice (Gp B vs Gp A: ascending aorta, ex vivo, 1.07 ± 0.21 mm vs 1.80 ± 0.67 mm [P < .05]; aortic arch, ex vivo, 1.51 ± 0.40 mm vs 2.70 ± 1.06 mm [P < .05]; Gp C vs Gp A: ascending aortas, ex vivo, 1.10 ± 0.33 mm vs 1.80 ± 0.67 mm [P < .05]; aortic arch, ex vivo, 1.55 ± 0.56 mm vs 2.70 ± 1.06 mm [P < .05]). TAAD mice exhibited elastin fragmentation, abundant inflammatory cell infiltration, and significantly increased matrix metalloproteinase production in the aorta, and rapamycin treatment alleviated these changes. The protein levels of p-S6K and p-S6 in TAAD aortic tissues increased significantly, whereas they were suppressed by rapamycin. Conclusions: Rapamycin suppressed TAAD formation, probably by inhibition of mechanistic target of rapamycin signaling and reduction of inflammatory cell infiltration and matrix metalloproteinase 9 production. Targeting of the mechanistic target of rapamycin signaling pathway using rapamycin may be a favorable modulation for the clinical treatment of TAAD.
-
Investigation of the Pathogenic Mechanism of Ciprofloxacin in Aortic Aneurysm and Dissection by an Integrated Proteomics and Network Pharmacology Strategy
Zhaoran Chen,Jianqiang Wu,Wei Wang,Xiaoyue Tang,Lei Zhou,Yanze Lv,Yuehong Zheng
DOI: https://doi.org/10.3390/jcm12041270
IF: 3.9
2023-02-07
Journal of Clinical Medicine
Abstract:Aortic aneurysm and dissection (AAD) is a life-threatening disease worldwide. Recently, fluoroquinolones have been reported to significantly increase the risk of AAD. This study aimed to investigate the potential functional mechanism and molecular targets of fluoroquinolones in relation to AAD by an integrated proteomic and network pharmacology strategy. A total of 1351 differentially expressed proteins were identified in human aortic vascular smooth muscle cells (VSMCs) after ciprofloxacin (CIP) stimulation. The functional analysis emphasized the important roles of metabolism, extracellular matrix homeostasis, mitochondrial damage, focal adhesion, and apoptosis in CIP-stimulated VSMCs. CIP targets were predicted with online databases and verified by molecular docking. Protein–protein interaction (PPI) analysis and module construction of the 34 potential CIP targets and 37 selected hub molecules after CIP stimulation identified four critical target proteins in the module: PARP1, RAC1, IGF1R and MKI67. Functional analysis of the PPI module showed that the MAPK signalling pathway, focal adhesion, apoptosis, regulation of actin cytoskeleton, and PI3K-Akt signalling pathway were significantly enriched. Our results will provide novel insights into the pathogenic mechanism of fluoroquinolones in aortic diseases.
medicine, general & internal
-
Ciprofloxacin Accelerates Angiotensin-II-Induced Vascular Smooth Muscle Cells Senescence Through Modulating AMPK/ROS pathway in Aortic Aneurysm and Dissection
Weiyue Zeng,Yaowen Liang,Shangjun Huang,Jiarui Zhang,Cong Mai,Binbin He,Linli Shi,Baojuan Liu,Weifeng Li,Xiaoran Huang,Xin Li
DOI: https://doi.org/10.1007/s12012-024-09892-z
Abstract:Aortic aneurysm and dissection (AAD) is a cardiovascular disease that poses a severe threat to life and has high morbidity and mortality rates. Clinical and animal-based studies have irrefutably shown that fluoroquinolones, a commonly prescribed antibiotic for treating infections, significantly increase the risk of AAD. Despite this, the precise mechanism by which fluoroquinolones cause AAD remains unclear. Therefore, this study aims to investigate the molecular mechanism and role of Ciprofloxacin definitively-a type of fluoroquinolone antibiotic-in the progression of AAD. Aortic transcriptome data were collected from GEO datasets to detect the genes and pathways expressed differently between healthy donors and AAD patients. Human primary Vascular Smooth Muscle Cells (VSMCs) were isolated from the aorta. After 72 h of exposure to 110ug/ml Ciprofloxacin or 100 nmol/L AngII, either or combined, the senescent cells were identified through SA-β-gal staining. MitoTracker staining was used to examine the morphology of mitochondria in each group. Cellular Reactive Oxygen Species (ROS) levels were measured using MitoSox and DCFH-DA staining. Western blot assay was performed to detect the protein expression level. We conducted an analysis of transcriptome data from both healthy donors and patients with AAD and found that there were significant changes in cellular senescence-related signaling pathways in the latter group. We then isolated and identified human primary VSMCs from healthy donors (control-VSMCs) and patients' (AAD-VSMCs) aortic tissue, respectively. We found that VSMCs from patients exhibited senescent phenotype as compared to control-VSMCs. The higher levels of p21 and p16 and elevated SA-β-gal activity demonstrated this. We also found that pretreatment with Ciprofloxacin promoted angiotensin-II-induced cellular senescence in control-VSMCs. This was evidenced by increased SA-β-gal activity, decreased cell proliferation, and elevation of p21 and p16 protein levels. Additionally, we found that Angiotensin-II (AngII) induced VSMC senescence by promoting ROS generation. We used DCFH-DA and mitoSOX staining to identify that Ciprofloxacin and AngII pretreatment further elevated ROS levels than the vehicle or alone group. Furthermore, JC-1 staining showed that mitochondrial membrane potential significantly declined in the Ciprofloxacin and AngII combination group compared to others. Compared to the other three groups, pretreatment of Ciprofloxacin plus AngII could further induce mitochondrial fission, demonstrated by mitoTracker staining and western blotting assay. Mechanistically, we found that Ciprofloxacin impaired the balance of mitochondrial fission and fusion dynamics in VSMCs by suppressing the phosphorylation of AMPK signaling. This caused mitochondrial dysfunction and ROS generation, thereby elevating AngII-induced cellular senescence. However, treatment with the AMPK activator partially alleviated those effects. Our data indicate that Ciprofloxacin may accelerate AngII-induced VSMC senescence through modulating AMPK/ROS signaling and, subsequently, hasten the progression of AAD.
-
Angiopoietin-like protein 8 deficiency attenuates thoracic aortic aneurysm/dissection development in β-aminopropionitrile monofumarate-induced model mice
Yun-Yun Yang,Xiao-Lu Jiao,Hua-Hui Yu,Lin-Yi Li,Juan Li,Xiao-Ping Zhang,Yan-Wen Qin
DOI: https://doi.org/10.1016/j.bbadis.2022.166619
Abstract:Thoracic aortic aneurysm/dissection (TAAD) is a life-threatening cardiovascular disorder. Endoplasmic reticulum stress (ERS) and vascular smooth muscle cell (VSMC) apoptosis are involved in TAAD progression. The Protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) pathway is associated with VSMC apoptosis. Serum Angiopoietin-Like Protein 8 (ANGPTL8) levels are associated with aortic diameter and rupture rate of TAAD. However, a direct role of ANGPTL8 in TAAD has not been determined. β-Aminopropionitrile monofumarate (BAPN) was used to induce TAAD in C57BL/6 mice. ANGPTL8 knockout mice were used to detect the effects of ANGPTL8 on TAAD development. ANGPTL8knockdown in vitro was used to analyze the role of ANGPTL8 in VSMCs and ERS. In addition, over-expression of ANGPTL8 in VSMCs and a PERK inhibitor were used to assess the effect of ANGPTL8 on the PERK pathway. ANGPTL8 levels were increased in the aortic wall and VSMCs of BAPN-induced TAAD mice. Compared with BAPN-treated wild-type mice, ANGPTL8 knockout significantly reduced the rupture rate of TAAD to 0 %. In addition, the protein levels of proinflammatory cytokines and matrix metalloproteinase 9 (MMP9) and ERS proteins were decreased in the aorta wall. Angptl8 shRNA decreased MMP9 and ERS protein levels in VSMCs in vitro. Overexpression of ANGPTL8 significantly increased the levels of ERS proteins and MMPs, while a PERK inhibitor significantly decreased the effects of ANGPTL8 in VSMCs. ANGPTL8 contributed to TAAD development by inducing ERS activation and degradation of extracellular matrix in the aorta wall. Inhibition of ANGPTL8 may therefore represent a new strategy for TAAD therapy.
-
Inhibition of heme oxygenase 1 alleviates thoracic aortic aneurysm via restoration of extracellular matrix
Wenyu Song,Kangjie Shen,Guangguo Fu,Lieyang Qin,Ghufran Bagaber,Jinmiao Chen,Lai Wei
DOI: https://doi.org/10.1016/j.bbrc.2023.149405
2024-01-29
Abstract:Background: Thoracic aortic aneurysm (TAA) is a silent but life-threatening cardiovascular disease. Heme oxygenase 1 (HO-1) plays an important role in the cardiovascular diseases but is poorly understood in TAA. This study aims at investigating the role of HO-1 in TAA. Methods: Single-cell RNA sequencing, Western blot and histological assay were performed to identify specific cellular expression of HO-1 in both human and β-aminopropionitrile (BAPN)-induced mice TAA. Zinc protoporphyrin (ZnPP), a pharmacological inhibitor of HO-1, was used to investigate whether inhibition of HO-1 could attenuate BAPN-induced TAA in rodent model. Histological assay, Western blot assay, and mRNA sequencing were further performed to explore the underlying mechanisms. Results: Single-cell transcriptomic analyses of 113,800 thoracic aortic cells identified an increase of HO-1(+) macrophage in aneurysmal thoracic aorta from BAPN-induced TAA mice and TAA patients. Histological assay verified HO-1 overexpression in clinical TAA specimens, which was co-localized with CD68(+) macrophage. HO-1(+) macrophage was closely associated with pro-inflammatory response and immune activation. Inhibition of HO-1 through ZnPP significantly alleviated BAPN-induced TAA in mice and restored extracellular matrix (ECM) in vivo. Further experiments showed that ZnPP treatment suppressed the expression of matrix metalloproteinases (MMPs) in aneurysmal thoracic aortic tissues from BAPN-induced TAA mice, including MMP2 and MMP9. Macrophages from myeloid specific HO-1 knockout mice displayed weakened pro-inflammatory activity and ECM degradation capability. Conclusion: HO-1(+) macrophage subgroup is a typical hallmark of TAA. Inhibition of HO-1 through ZnPP alleviates BAPN-induced TAA in mice, which might work through restoration of ECM via suppressing MMP2 and MMP9 expression.
-
Blocking Interleukin-1 Beta Reduces the Evolution of Thoracic Aortic Dissection in a Rodent Model
Ling-Ling Guo,Meng-Tao Wu,Li-Wei Zhang,Yong-Xin Chu,Peng Tian,Zai-Ping Jing,Jia-Si Li,Yu-Dong Sun,Kak K Yeung,Lei Zhang,Kak K. Yeung
DOI: https://doi.org/10.1016/j.ejvs.2020.08.032
IF: 6.427
2020-12-01
European Journal of Vascular and Endovascular Surgery
Abstract:<h3 class="u-h4 u-margin-m-top u-margin-xs-bottom">Objective</h3><p>Thoracic aortic dissection (TAD) is associated with matrix changes, biochemical changes, and inflammatory markers like interleukin-1 beta (IL-1β). However, the exact mechanism remains unknown. This study aimed to investigate the role of IL-1β, matrix metalloproteinase (MMP)-2, MMP-9, smooth muscle cell apoptosis, and elastic fibre fracture in the development of TAD in a rat model.</p><h3 class="u-h4 u-margin-m-top u-margin-xs-bottom">Methods</h3><p>The TAD rat model was induced by β-aminopropionitrile (BAPN). TAD was investigated in 112 male Sprague–Dawley rats, which were equally divided into four groups of 28 rats (Control, BAPN, BAPN + IL-1β, and BAPN + IL-1β antibody). Systolic blood pressure, survival, and the development of TAD were measured after six weeks. Expression of IL-1β, MMP-2, and MMP-9 was measured by Western blot. Apoptosis, aortic elastin concentration, and biomechanical characteristics were measured by the TdT mediated dUTP nick end labelling assay, Victoria blue staining, and <em>in vitro</em> testing.</p><h3 class="u-h4 u-margin-m-top u-margin-xs-bottom">Results</h3><p>During six weeks, the mortality was 0% (0/28) in the control group, 53.6% (15/28) in the BAPN group (<em>p</em> < .001 compared with the control group), 75.0% (21/28) in the BAPN + IL-1β group (<em>p</em> = .007 compared with the BAPN group), and 35.7% (10/28) in the BAPN + IL-1β antibody group (<em>p</em> = .023 compared with BAPN group and <em>p</em> < .001 compared with the BAPN + IL-1β group). IL-1β treatment deteriorates BAPN induced mortality and aneurysm expansion, which were attenuated by anti-IL-1β treatment. In BAPN + IL-1β group, stress and strain parameters were decreased by 13.5%–53.5% and elastin content was decreased by 14%, and IL-1β, MMP-2, and MMP-9 were expressed higher by 117%, 108%, and 75% when compared with the rats in the BAPN group. Contrarily, in the BAPN + IL-1β antibody group, the above changes could be completely (strain, elastin content, and expression of MMP-2) or partly (elasticity modulus, stress, and expression of MMP-9) blocked by anti-IL-1β treatment.</p><h3 class="u-h4 u-margin-m-top u-margin-xs-bottom">Conclusion</h3><p>IL-1β plays a critical role in TAD formation by altering the expression of MMP-2 and MMP-9, degrading the aortic wall matrix, causing elastic fibre rupture, and changing the stress or strain of the aortic wall. Anti-IL-1β reduces the later effects and could be one of the molecular targets for prognosis and drug treatment of TAD in the future.</p>
surgery,peripheral vascular disease