1
|
Ke ZX, Chen GZ, Hu K, Zhang S, Zhou P, Chen DX, Li YQ, Li Q, Yang C. Safety and Efficacy of Endovascular Aortic Repair for Abdominal Aortic Aneurysms with a Hostile Neck Anatomy. Curr Med Sci 2023; 43:1221-1228. [PMID: 38153630 DOI: 10.1007/s11596-023-2822-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 01/14/2023] [Indexed: 12/29/2023]
Abstract
OBJECTIVE This study aimed to investigate the safety and efficacy of endovascular aortic repair (EVAR) for the treatment of an abdominal aortic aneurysm (AAA) with a hostile neck anatomy (HNA). METHODS From January 1, 2015 to December 31, 2019, a total of 259 patients diagnosed with an AAA who underwent EVAR were recruited into this study. Based on the morphological characteristics of the proximal neck anatomy, the patients were divided into the HNA group and the friendly neck anatomy (FNA) group. The patients were followed up for up to 4 years. RESULTS The average follow-up time was 1056.1±535.5 days. Type I endoleak occurred in 4 patients in the HNA group, and 2 patients in the FNA group. Neither death nor intraoperative switch to open repair occurred in either group. The time of the operation was significantly longer in the HNA group (FNA vs. HNA, 99.2±51.1 min vs. 117.5±63.8 min, P=0.011). There were no significant differences in short-term clinical success rate (P=0.228) or midterm clinical success rate (P=0.889) between the two groups. The overall mortality rate was 10.4%, and Kaplan-Meier survival analysis indicated that the two groups had similar cumulative survival rates at the end of the follow-up period (P=0.889). CONCLUSION EVAR was feasible and safe in patients with an AAA with a proximal HNA. The early and midterm results were promising; however, further studies are needed to verify the long-term effectiveness of EVAR.
Collapse
Affiliation(s)
- Zun-Xiang Ke
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ge-Zheng Chen
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ke Hu
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shan Zhang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Peng Zhou
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dian-Xi Chen
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yi-Qing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qin Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Chao Yang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
2
|
Yu Y, Wu XQ, Su FF, Yue CF, Zhou XM, Xu C. Maximakinin reduced intracellular Ca 2+ level in vascular smooth muscle cells through AMPK/ERK1/2 signaling pathways. Hypertens Res 2023; 46:1949-1960. [PMID: 37258626 DOI: 10.1038/s41440-023-01330-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 05/09/2023] [Accepted: 05/13/2023] [Indexed: 06/02/2023]
Abstract
We detect the antihypertensive effects of maximakinin (MK) on renal hypertensive rats (RHRs) and further research the influence of MK on vascular smooth muscle cells (VSMCs) to explore its hypotensive mechanism. The effects of MK on arterial blood pressure were observed in RHRs. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide (MTT) assays were performed to detect the effect of MK on VSMC viability. Western blot and flow cytometry were used to investigate the influence of MK on intracellular Ca2+ levels and protein expression changes in VSMCs. In addition, specific protein inhibitors were applied to confirm the involvement of Ca2+-related signaling pathways induced by MK in VSMCs. MK showed a more significant antihypertensive effect than bradykinin in RHRs. MK significantly decreased intracellular Ca2+ concentrations. Furthermore, MK significantly induced the phosphorylation of signaling molecules, including extracellular signal-regulated kinase 1/2 (ERK1/2), P38, AMP-activated protein kinase (AMPK) and Akt in VSMCs. Moreover, only ERK1/2 inhibitor U0126 and AMPK inhibitor Compound C completely restored the decreased intracellular Ca2+ level induced by MK, and further research demonstrated that AMPK functioned upstream of ERK1/2 following exposure to MK. Finally, HOE-140, an inhibitor of the bradykinin B2 receptors (B2Rs), was applied to investigate the potential targets of MK in VSMCs. HOE-140 significantly blocked the AMPK/ERK1/2 pathway induced by MK, suggesting that the B2Rs might play an important role in MK-induced AMPK and ERK1/2 activation. MK significantly reduces blood pressure in RHRs. MK exerts its antihypertensive effect by activating the B2Rs and downstream AMPK/ERK1/2 pathways, leading to significantly reduced Ca2+ levels in VSMCs.
Collapse
Affiliation(s)
- Yang Yu
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe, Shenyang, Liaoning, 110016, China
| | - Xue-Qian Wu
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe, Shenyang, Liaoning, 110016, China
| | - Fan-Fan Su
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe, Shenyang, Liaoning, 110016, China
| | - Cai-Feng Yue
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe, Shenyang, Liaoning, 110016, China
| | - Xiao-Mian Zhou
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe, Shenyang, Liaoning, 110016, China
| | - Cheng Xu
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
3
|
Li FXZ, Liu JJ, Xu F, Shan SK, Zheng MH, Lei LM, Lin X, Guo B, Li CC, Wu F, Tang KX, Cao YC, Wu YY, Duan JY, Wu YL, He SY, Chen X, Yuan LQ. Cold exposure protects against medial arterial calcification development via autophagy. J Nanobiotechnology 2023; 21:226. [PMID: 37461031 PMCID: PMC10351118 DOI: 10.1186/s12951-023-01985-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023] Open
Abstract
Medial arterial calcification (MAC), a systemic vascular disease different from atherosclerosis, is associated with an increased incidence of cardiovascular events. Several studies have demonstrated that ambient temperature is one of the most important factors affecting cardiovascular events. However, there has been limited research on the effect of different ambient temperatures on MAC. In the present study, we showed that cold temperature exposure (CT) in mice slowed down the formation of vitamin D (VD)-induced vascular calcification compared with room temperature exposure (RT). To investigate the mechanism involved, we isolated plasma-derived exosomes from mice subjected to CT or RT for 30 days (CT-Exo or RT-Exo, respectively). Compared with RT-Exo, CT-Exo remarkably alleviated the calcification/senescence formation of vascular smooth muscle cells (VSMCs) and promoted autophagy by activating the phosphorylation of AMP-activated protein kinase (p-AMPK) and inhibiting phosphorylation of mammalian target of rapamycin (p-mTOR). At the same time, CT-Exo promoted autophagy in β-glycerophosphate (β-GP)-induced VSMCs. The number of autophagosomes and the expression of autophagy-related proteins ATG5 and LC3B increased, while the expression of p62 decreased. Based on a microRNA chip microarray assay and real-time polymerase chain reaction, miR-320a-3p was highly enriched in CT-Exo as well as thoracic aortic vessels in CT mice. miR-320a-3p downregulation in CT-Exo using AntagomiR-320a-3p inhibited autophagy and blunted its anti-calcification protective effect on VSMCs. Moreover, we identified that programmed cell death 4 (PDCD4) is a target of miR-320a-3p, and silencing PDCD4 increased autophagy and decreased calcification in VSMCs. Treatment with CT-Exo alleviated the formation of MAC in VD-treated mice, while these effects were partially reversed by GW4869. Furthermore, the anti-arterial calcification protective effects of CT-Exo were largely abolished by AntagomiR-320a-3p in VD-induced mice. In summary, we have highlighted that prolonged cold may be a good way to reduce the incidence of MAC. Specifically, miR-320a-3p from CT-Exo could protect against the initiation and progression of MAC via the AMPK/mTOR autophagy pathway.
Collapse
Affiliation(s)
- Fu-Xing-Zi Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Jun-Jie Liu
- Department of Periodontal Division, Hunan Xiangya Stomatological Hospital, Central South University, Changsha, China
| | - Feng Xu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Su-Kang Shan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Ming-Hui Zheng
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Li-Min Lei
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Xiao Lin
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bei Guo
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Chang-Chun Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Feng Wu
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ke-Xin Tang
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Ye-Chi Cao
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yun-Yun Wu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Jia-Yue Duan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yan-Lin Wu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Si-Yang He
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Xi Chen
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Ling-Qing Yuan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
4
|
Fang ZM, Feng X, Chen Y, Luo H, Jiang DS, Yi X. Targeting autophagy in aortic aneurysm and dissection. Biomed Pharmacother 2022; 153:113547. [PMID: 36076620 DOI: 10.1016/j.biopha.2022.113547] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/03/2022] [Accepted: 08/11/2022] [Indexed: 01/18/2023] Open
|
5
|
Chen CH, Ho HH, Jiang WC, Ao-Ieong WS, Wang J, Orekhov AN, Sobenin IA, Layne MD, Yet SF. Cysteine-rich protein 2 deficiency attenuates angiotensin II-induced abdominal aortic aneurysm formation in mice. J Biomed Sci 2022; 29:25. [PMID: 35414069 PMCID: PMC9004090 DOI: 10.1186/s12929-022-00808-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 04/01/2022] [Indexed: 11/10/2022] Open
Abstract
Background Abdominal aortic aneurysm (AAA) is a relatively common and often fatal condition. A major histopathological hallmark of AAA is the severe degeneration of aortic media with loss of vascular smooth muscle cells (VSMCs), which are the main source of extracellular matrix (ECM) proteins. VSMCs and ECM homeostasis are essential in maintaining structural integrity of the aorta. Cysteine-rich protein 2 (CRP2) is a VSMC-expressed protein; however, the role of CRP2 in AAA formation is unclear. Methods To investigate the function of CRP2 in AAA formation, mice deficient in Apoe (Apoe−/−) or both CRP2 (gene name Csrp2) and Apoe (Csrp2−/−Apoe−/−) were subjected to an angiotensin II (Ang II) infusion model of AAA formation. Aortas were harvested at different time points and histological analysis was performed. Primary VSMCs were generated from Apoe−/− and Csrp2−/−Apoe−/− mouse aortas for in vitro mechanistic studies. Results Loss of CRP2 attenuated Ang II-induced AAA incidence and severity, accompanied by preserved smooth muscle α-actin expression and reduced elastin degradation, matrix metalloproteinase 2 (MMP2) activity, deposition of collagen, particularly collagen III (Col III), aortic tensile strength, and blood pressure. CRP2 deficiency decreased the baseline MMP2 and Col III expression in VSMCs and mitigated Ang II-induced increases of MMP2 and Col III via blunting Erk1/2 signaling. Rescue experiments were performed by reintroducing CRP2 into Csrp2−/−Apoe−/− VSMCs restored Ang II-induced Erk1/2 activation, MMP2 expression and activity, and Col III levels. Conclusions Our results indicate that in response to Ang II stimulation, CRP2 deficiency maintains aortic VSMC density, ECM homeostasis, and structural integrity through Erk1/2–Col III and MMP2 axis and reduces AAA formation. Thus, targeting CRP2 provides a potential therapeutic strategy for AAA. Supplementary information The online version contains supplementary material available at 10.1186/s12929-022-00808-z.
Collapse
Affiliation(s)
- Chung-Huang Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, 35053, Zhunan, Taiwan
| | - Hua-Hui Ho
- Institute of Cellular and System Medicine, National Health Research Institutes, 35053, Zhunan, Taiwan
| | - Wei-Cheng Jiang
- Institute of Cellular and System Medicine, National Health Research Institutes, 35053, Zhunan, Taiwan
| | - Wai-Sam Ao-Ieong
- Department of Chemical Engineering, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Jane Wang
- Department of Chemical Engineering, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | | | - Igor A Sobenin
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, 121552, Moscow, Russia
| | - Matthew D Layne
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Shaw-Fang Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, 35053, Zhunan, Taiwan. .,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
6
|
He C, Jiang B, Wang M, Ren P, Murtada SI, Caulk AW, Li G, Qin L, Assi R, Lovoulos CJ, Schwartz MA, Humphrey JD, Tellides G. mTOR inhibition prevents angiotensin II-induced aortic rupture and pseudoaneurysm but promotes dissection in Apoe-deficient mice. JCI Insight 2022; 7:155815. [PMID: 35132962 PMCID: PMC8855820 DOI: 10.1172/jci.insight.155815] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/23/2021] [Indexed: 01/04/2023] Open
Abstract
Aortic dissection and rupture are triggered by decreased vascular wall strength and/or increased mechanical loads. We investigated the role of mTOR signaling in aortopathy using a well-described model of angiotensin II–induced dissection, aneurysm, or rupture of the suprarenal abdominal aorta in Apoe-deficient mice. Although not widely appreciated, nonlethal hemorrhagic lesions present as pseudoaneurysms without significant dissection in this model. Angiotensin II–induced aortic tears result in free rupture, contained rupture with subadventitial hematoma (forming pseudoaneurysms), dilatation, or healing, while the media invariably thickens regardless of mural tears. Medial thickening results from smooth muscle cell hypertrophy and extracellular matrix accumulation, including matricellular proteins. Angiotensin II activates mTOR signaling in vascular wall cells, and inhibition of mTOR signaling by rapamycin prevents aortic rupture but promotes dissection. Decreased aortic rupture correlates with decreased inflammation and metalloproteinase expression, whereas extensive dissection correlates with induction of matricellular proteins that modulate adhesion of vascular cells. Thus, mTOR activation in vascular wall cells determines whether aortic tears progress to dissection or rupture. Previous mechanistic studies of aortic aneurysm and dissection by angiotensin II in Apoe-deficient mice should be reinterpreted as clinically relevant to pseudoaneurysms, and mTOR inhibition for aortic disease should be explored with caution.
Collapse
Affiliation(s)
- Changshun He
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Bo Jiang
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Mo Wang
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Pengwei Ren
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Sae-Il Murtada
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut, USA
| | - Alexander W Caulk
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut, USA
| | - Guangxin Li
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Lingfeng Qin
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Roland Assi
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA.,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA.,Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Constantinos J Lovoulos
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA.,Department of Surgery, Frank H. Netter MD School of Medicine, Quinnipiac University, North Haven, Connecticut, USA
| | - Martin A Schwartz
- Department of Medicine (Cardiology).,Department of Cell Biology, and.,Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut, USA.,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA
| | - George Tellides
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA.,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA.,Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| |
Collapse
|
7
|
Estrada AC, Irons L, Rego BV, Li G, Tellides G, Humphrey JD. Roles of mTOR in thoracic aortopathy understood by complex intracellular signaling interactions. PLoS Comput Biol 2021; 17:e1009683. [PMID: 34898595 PMCID: PMC8700007 DOI: 10.1371/journal.pcbi.1009683] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/23/2021] [Accepted: 11/26/2021] [Indexed: 02/01/2023] Open
Abstract
Thoracic aortopathy–aneurysm, dissection, and rupture–is increasingly responsible for significant morbidity and mortality. Advances in medical genetics and imaging have improved diagnosis and thus enabled earlier prophylactic surgical intervention in many cases. There remains a pressing need, however, to understand better the underlying molecular and cellular mechanisms with the hope of finding robust pharmacotherapies. Diverse studies in patients and mouse models of aortopathy have revealed critical changes in multiple smooth muscle cell signaling pathways that associate with disease, yet integrating information across studies and models has remained challenging. We present a new quantitative network model that includes many of the key smooth muscle cell signaling pathways and validate the model using a detailed data set that focuses on hyperactivation of the mechanistic target of rapamycin (mTOR) pathway and its inhibition using rapamycin. We show that the model can be parameterized to capture the primary experimental findings both qualitatively and quantitatively. We further show that simulating a population of cells by varying receptor reaction weights leads to distinct proteomic clusters within the population, and that these clusters emerge due to a bistable switch driven by positive feedback in the PI3K/AKT/mTOR signaling pathway. Cell signaling drives changes across scales, from altered transcription at the single-cell level to tissue-level growth and remodeling. Studying complex interactions within cell signaling pathways can lead to a better understanding of the progression of disease. In particular, we are interested in how vascular cells can change their phenotype in a way that exacerbates aortopathy, namely, the development of aneurysms, dissections, and rupture. In this study we built a novel cell signaling network model of a vascular smooth muscle cell using archival data and used it to capture the effects of a genetic knock-out and subsequent pharmacologic rescue. We then used the model to simulate populations of smooth muscle cells and found that small perturbations to the strength of signaling can lead to distinct clusters of cells. With further analysis of the network substructures, we found that a positive feedback loop within the network was responsible for the distinct phenotypes we saw in our clusters of simulated cells. We believe that this work not only helps us to understand changes in smooth muscle cell phenotype but also opens the possibility to study other signaling perturbations associated with aortopathy.
Collapse
Affiliation(s)
- Ana C. Estrada
- Department of Biomedical Engineering, Yale University; New Haven, Connecticut, United States of America
| | - Linda Irons
- Department of Biomedical Engineering, Yale University; New Haven, Connecticut, United States of America
| | - Bruno V. Rego
- Department of Biomedical Engineering, Yale University; New Haven, Connecticut, United States of America
| | - Guangxin Li
- Department of Surgery, Yale School of Medicine; New Haven, Connecticut, United States of America
| | - George Tellides
- Department of Surgery, Yale School of Medicine; New Haven, Connecticut, United States of America
- Vascular Biology and Therapeutics Program, Yale School of Medicine; New Haven, Connecticut, United States of America
| | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University; New Haven, Connecticut, United States of America
- Vascular Biology and Therapeutics Program, Yale School of Medicine; New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
8
|
Zhou Z, Zhou H, Zou X, Wang X. RUNX3 is up-regulated in abdominal aortic aneurysm and regulates the function of vascular smooth muscle cells by regulating TGF-β1. J Mol Histol 2021; 53:1-11. [PMID: 34813022 DOI: 10.1007/s10735-021-10035-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 10/23/2021] [Indexed: 02/06/2023]
Abstract
Abdominal aortic aneurysm (AAA) has been associated with the dysfunction of vascular smooth muscle cells (VSMCs) and extracellular matrix (ECM) remodelling. Runt-related transcription factor 3 (RUNX3) has been reported to be up-regulated in aneurysmal aorta samples compared with normal aorta. However, its function in VSMCs and the mechanism of function remains unknown. Therefore, our study aimed to investigate the role of RUNX3 in ECM remodelling and VSMC function, and further explore the underlying mechanism. Our results verified that RUNX3 was increased in aortic samples of AAA compared with healthy controls. Overexpression vectors of RUNX3 (ov-RUNX3) and siRNA of RUNX3 (si-RUNX3) were transfected into Human aortic smooth muscle cells (HAoSMCs). The results indicated that ov-RUNX3 promoted cell proliferation, migration, and MMP-2/3/9 secretion, and suppressed TIMP-1, collagen I/III, SM22, MYH11 and CNN1 expression in HAoSMCs. The silencing of RUNX3 has the opposite effect. Furthermore, we found that RUNX3 targets TGF-β1 and suppressed its transcription. The silencing of TGF-β1 increased cell proliferation, migration and MMP-2/3/9 expression, and inhibited TIMP-1, Collagen I/III, SM22, MYH11 and CNN1 expression. In addition, TGF-β1 reversed the effect of RUNX3 overexpression on HAoSMCs. Hence, our study indicated that RUNX3 promotes cell proliferation, migration, and ECM remodelling through suppressing TGF-β1.
Collapse
Affiliation(s)
- Zhongxiao Zhou
- Department of Vascular Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, No. 70 Heping Road, Huancui District, Weihai, 264200, China
| | - Haimeng Zhou
- Department of Vascular Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, No. 70 Heping Road, Huancui District, Weihai, 264200, China.
| | - Xin Zou
- Department of Vascular Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, No. 70 Heping Road, Huancui District, Weihai, 264200, China
| | - Xiaowei Wang
- Department of Vascular Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, No. 70 Heping Road, Huancui District, Weihai, 264200, China
| |
Collapse
|
9
|
Xiong JM, Liu H, Chen J, Zou QQ, Wang YYJ, Bi GS. Curcumin nicotinate suppresses abdominal aortic aneurysm pyroptosis via lncRNA PVT1/miR-26a/KLF4 axis through regulating the PI3K/AKT signaling pathway. Toxicol Res (Camb) 2021; 10:651-661. [PMID: 34141179 DOI: 10.1093/toxres/tfab041] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/19/2021] [Accepted: 04/08/2021] [Indexed: 11/15/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a chronic dilated disease of the aorta that is characterized by chronic inflammation. Curcumin (Cur) is previously showed to attenuate AAA by inhibiting inflammatory response in ApoE -/- mice. Since Cur has the limitations of aqueous solubility and instability. Here, we focus on the role of curcumin nicotinate (CurTn), a Cur derivative is derived from Cur and nicotinate. An in vitro model of AAA was established by treating vascular smooth muscle cells (VSMCs) with II (Ang-II). Gene and protein expressions were examined by quantitative real-time PCR (qPCR) or western blotting. Cell migration and pyroptosis were determined by transwell assay and flow cytometry. The interaction between plasmacytoma variant translocation 1 (PVT1), miR-26a and krüppel-like factor 4 (KLF4) was predicted by online prediction tool and confirmed by luciferase reporter assay. CurTn reduced Ang-II-induced AAA-associated proteins, inflammatory cytokine expressions, and attenuated pyroptosis in VSMCs. PVT1 overexpression suppressed the inhibitory effect of CurTn on AngII-induced pyroptosis and inflammatory in VSMCs by sponging miR-26a. miR-26a directly targeted KLF4 and suppressed its expression, which eventually led to the deactivation of the PI3K/AKT signaling pathway. Besides, the regulatory effect of CurTn on pyroptosis of VSMCs induced by Ang-II was reversed through the PVT1/miR-26a/KLF4 pathway. In short, CurTn suppressed VSMCs pyroptosis and inflammation though mediation PVT1/miR-26a/KLF4 axis by regulating the PI3K/AKT signaling pathway, CurTn might consider as a potential therapeutic target in the treatment of AAA.
Collapse
Affiliation(s)
- Jian-Ming Xiong
- Department of Vascular Surgery, Yiyang Central Hospital, Yiyang 413000, Hunan Province, P.R. China
| | - Hui Liu
- Department of Vascular Surgery, Yiyang Central Hospital, Yiyang 413000, Hunan Province, P.R. China
| | - Jie Chen
- Department of Vascular Surgery, The Second Affiliated Hospital, University of South China, Hengyang 421000, Hunan Province, P.R. China
| | - Qing-Qing Zou
- Department of Vascular Surgery, The Second Affiliated Hospital, University of South China, Hengyang 421000, Hunan Province, P.R. China
| | - Yang-Yi-Jing Wang
- Department of Vascular Surgery, The Second Affiliated Hospital, University of South China, Hengyang 421000, Hunan Province, P.R. China
| | - Guo-Shan Bi
- Department of Vascular Surgery, The Second Affiliated Hospital, University of South China, Hengyang 421000, Hunan Province, P.R. China
| |
Collapse
|
10
|
Zhang Y, Wang Y, Zhang L, Xia L, Zheng M, Zeng Z, Liu Y, Yarovinsky T, Ostriker AC, Fan X, Weng K, Su M, Huang P, Martin KA, Hwa J, Tang WH. Reduced Platelet miR-223 Induction in Kawasaki Disease Leads to Severe Coronary Artery Pathology Through a miR-223/PDGFRβ Vascular Smooth Muscle Cell Axis. Circ Res 2020; 127:855-873. [PMID: 32597702 PMCID: PMC7486265 DOI: 10.1161/circresaha.120.316951] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/26/2020] [Indexed: 12/13/2022]
Abstract
RATIONALE Kawasaki disease (KD) is an acute vasculitis of early childhood that can result in permanent coronary artery structural damage. The cause for this arterial vulnerability in up to 15% of patients with KD is unknown. Vascular smooth muscle cell dedifferentiation play a key role in the pathophysiology of medial damage and aneurysm formation, recognized arterial pathology in KD. Platelet hyperreactivity is also a hallmark of KD. We recently demonstrated that uptake of platelets and platelet-derived miRNAs influences vascular smooth muscle cell phenotype in vivo. OBJECTIVE We set out to explore whether platelet/vascular smooth muscle cell (VSMC) interactions contribute to coronary pathology in KD. METHODS AND RESULTS We prospectively recruited and studied 242 patients with KD, 75 of whom had documented coronary artery pathology. Genome-wide miRNA sequencing and droplet digital PCR demonstrated that patient with KD platelets have significant induction of miR-223 compared with healthy controls (HCs). Platelet-derived miR-223 has recently been shown to promote vascular smooth muscle quiescence and resolution of wound healing after vessel injury. Paradoxically, patients with KD with the most severe coronary pathology (giant coronary artery aneurysms) exhibited a lack of miR-223 induction. Hyperactive platelets isolated from patients with KD are readily taken up by VSMCs, delivering functional miR-223 into the VSMCs promoting VSMC differentiation via downregulation of PDGFRβ (platelet-derived growth factor receptor β). The lack of miR-223 induction in patients with severe coronary pathology leads to persistent VSMC dedifferentiation. In a mouse model of KD (Lactobacillus casei cell wall extract injection), miR-223 knockout mice exhibited increased medial thickening, loss of contractile VSMCs in the media, and fragmentation of medial elastic fibers compared with WT mice, which demonstrated significant miR-223 induction upon Lactobacillus casei cell wall extract challenge. The excessive arterial damage in the miR-223 knockout could be rescued by adoptive transfer of platelet, administration of miR-223 mimics, or the PDGFRβ inhibitor imatinib mesylate. Interestingly, miR-223 levels progressively increase with age, with the lowest levels found in <5-year-old. This provides a basis for coronary pathology susceptibility in this very young cohort. CONCLUSIONS Platelet-derived miR-223 (through PDGFRβ inhibition) promotes VSMC differentiation and resolution of KD induced vascular injury. Lack of miR-223 induction leads to severe coronary pathology characterized by VSMC dedifferentiation and medial damage. Detection of platelet-derived miR-223 in patients with KD (at the time of diagnosis) may identify patients at greatest risk of coronary artery pathology. Moreover, targeting platelet miR-223 or VSMC PDGFRβ represents potential therapeutic strategies to alleviate coronary pathology in KD. Graphic Abstract: A graphic abstract is available for this article.
Collapse
MESH Headings
- Adult
- Age Factors
- Animals
- Blood Platelets/metabolism
- Case-Control Studies
- Cells, Cultured
- Child
- Child, Preschool
- Coronary Artery Disease/etiology
- Coronary Artery Disease/genetics
- Coronary Artery Disease/metabolism
- Coronary Artery Disease/pathology
- Coronary Vessels/metabolism
- Coronary Vessels/pathology
- Disease Models, Animal
- Female
- Humans
- Infant
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- MicroRNAs/blood
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Mucocutaneous Lymph Node Syndrome/blood
- Mucocutaneous Lymph Node Syndrome/complications
- Mucocutaneous Lymph Node Syndrome/diagnosis
- Mucocutaneous Lymph Node Syndrome/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Platelet Activation
- Prospective Studies
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Severity of Illness Index
- Signal Transduction
- Young Adult
Collapse
Affiliation(s)
- Yuan Zhang
- From the Institute of Pediatrics (Y.Z., L.X., M.Z., Z.Z., Y.L., X.F., K.W., M.S., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Yanfei Wang
- Department of Cardiology (Y.W., L.Z., P.H.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Li Zhang
- Department of Cardiology (Y.W., L.Z., P.H.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Luoxing Xia
- From the Institute of Pediatrics (Y.Z., L.X., M.Z., Z.Z., Y.L., X.F., K.W., M.S., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Minhui Zheng
- From the Institute of Pediatrics (Y.Z., L.X., M.Z., Z.Z., Y.L., X.F., K.W., M.S., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Zhi Zeng
- From the Institute of Pediatrics (Y.Z., L.X., M.Z., Z.Z., Y.L., X.F., K.W., M.S., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Yingying Liu
- From the Institute of Pediatrics (Y.Z., L.X., M.Z., Z.Z., Y.L., X.F., K.W., M.S., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Timur Yarovinsky
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (T.Y., A.C.O., K.A.M., J.H.)
| | - Allison C Ostriker
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (T.Y., A.C.O., K.A.M., J.H.)
| | - Xuejiao Fan
- From the Institute of Pediatrics (Y.Z., L.X., M.Z., Z.Z., Y.L., X.F., K.W., M.S., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Kai Weng
- From the Institute of Pediatrics (Y.Z., L.X., M.Z., Z.Z., Y.L., X.F., K.W., M.S., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Meiling Su
- From the Institute of Pediatrics (Y.Z., L.X., M.Z., Z.Z., Y.L., X.F., K.W., M.S., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Ping Huang
- Department of Cardiology (Y.W., L.Z., P.H.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| | - Kathleen A Martin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (T.Y., A.C.O., K.A.M., J.H.)
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (T.Y., A.C.O., K.A.M., J.H.)
| | - Wai Ho Tang
- From the Institute of Pediatrics (Y.Z., L.X., M.Z., Z.Z., Y.L., X.F., K.W., M.S., W.H.T.), Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, China
| |
Collapse
|
11
|
Farrerol maintains the contractile phenotype of VSMCs via inactivating the extracellular signal-regulated protein kinase 1/2 and p38 mitogen-activated protein kinase signaling. Mol Cell Biochem 2020; 475:249-260. [PMID: 32840737 DOI: 10.1007/s11010-020-03878-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 08/07/2020] [Indexed: 12/17/2022]
Abstract
Farrerol, a dihydroflavone isolated from Rhododendron dauricum L., can inhibit vascular smooth muscle cell (VSMC) proliferation and exert a protective effect on H2O2-induced vascular endothelial cells injury. In this study, we investigated the effects of farrerol on VSMC phenotypic modulation and balloon injury-induced vascular neointimal formation and explored the underlying mechanisms. Serum-starved rat thoracic aorta SMCs (RASMCs) were first pretreated with farrerol (3, 10, and 30 μM, respectively), U0126 (a MEK kinase inhibitor), and SB203580 (a p38 kinase inhibitor), and followed by treatment with serum (10% FBS). The expression of several VSMC-specific markers, including α-SMA, SM22α, and OPN, were analyzed by western blot. Phosphorylation of extracellular signal-regulated protein kinase 1/2 (ERK 1/2) and p38 mitogen-activated protein kinase (MAPK) was also investigated. Farrerol inhibited the serum-induced transition of RASMCs from the contractile to the synthetic phenotype, and this was associated with a decrease in α-SMA and SM22α expression, and an increase in OPN expression. Farrerol also inhibited serum-induced phosphorylation of ERK1/2 and p38MAPK in RASMCs. Moreover, U0126 and SB203580 both inhibited the serum-induced phenotypic transition of RASMCs. These findings indicate that farrerol can maintain the contractile phenotype of VSMCs partly via inactivating the ERK1/2 and p38 MAPK signaling pathways. Using a rat model of carotid artery balloon injury, inhibition of VSMC phenotypic transition and suppression of neointimal formation were confirmed in vivo following the perivascular application of farrerol. Our results suggested that farrerol could be a promising lead compound for the treatment of vascular proliferative diseases.
Collapse
|
12
|
Therapeutic Effect of Rapamycin on Aortic Dissection in Mice. Int J Mol Sci 2020; 21:ijms21093341. [PMID: 32397282 PMCID: PMC7246910 DOI: 10.3390/ijms21093341] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 01/08/2023] Open
Abstract
Aortic dissection (AD) is a serious clinical condition that is unpredictable and frequently results in fatal outcome. Although rapamycin, an inhibitor of mechanistic target of rapamycin (mTOR), has been reported to be effective in preventing aortopathies in mouse models, its mode of action has yet to be clarified. A mouse AD model that was created by the simultaneous administration of β-aminopropionitrile (BAPN) and angiotensin II (AngII) for 14 days. Rapamycin treatment was started either at day 1 or at day 7 of BAPN+AngII challenge, and continued throughout the observational period. Rapamycin was effective both in preventing AD development and in suppressing AD progression. On the other hand, gefitinib, an inhibitor of growth factor signaling, did not show such a beneficial effect, even though both rapamycin and gefitinib suppressed cell cycle activation in AD. Rapamycin suppressed cell cycle-related genes and induced muscle development-related genes in an AD-related gene expression network without a major impact on inflammation-related genes. Rapamycin augmented the activation of Akt1, Akt2, and Stat3, and maintained the contractile phenotype of aortic smooth muscle cells. These findings indicate that rapamycin was effective both in preventing the development and in suppressing the progression of AD, indicating the importance of the mTOR pathway in AD pathogenesis.
Collapse
|
13
|
Chen DR, Jiang H, Chen J, Ruan CC, Han WQ, Gao PJ. Involvement of Angiotensin II Type 1 Receptor and Calcium Channel in Vascular Remodeling and Endothelial Dysfunction in Rats with Pressure Overload. Curr Med Sci 2020; 40:320-326. [PMID: 32337692 DOI: 10.1007/s11596-020-2171-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 03/19/2020] [Indexed: 01/03/2023]
Abstract
Vascular remodeling is an adaptive response to various stimuli, including mechanical forces, inflammatory cytokines and hormones. In the present study, we investigated the role of angiotensin II type 1 receptor (AT1R) and calcium channel in carotid artery remodeling in response to increased biomechanical forces by using the transverse aortic constriction (TAC) rat model. TAC was induced on ten-week-old male Sprague-Dawley rats and these models were treated with AT1R blocker olmesartan (1 mg/kg/day) or/and calcium channel blocker (CCB) amlodipine (0.5 mg/kg/day) for 14 days. After the treatment, the right common carotid artery proximal to the band (RCCA-B) was collected for further assay. Results showed that olmesartan, but not amlodipine, significantly prevented TAC-induced adventitial hyperplasia. Similarly, olmesartan, but not amlodipine, signifcantly prevented vascular infammation, as indicated by increased tumor necrosis factor α (TNF-α) and increased p65 phosphorylation, an indicator of nuclear factor κ-light-chain-enhancer of activated B cells (NFκB) activation in RCCA-B. In contrast, both olmesartan and amlodipine reversed the decreased expression of endothelial nitric oxidase synthase (eNOS) and improved endothelium-dependent vasodilation, whereas combination of olmesartan and amlodipine showed no further synergistic protective effects. These results suggest that AT1R was involved in vascular remodeling and inflammation in response to pressure overload, whereas AT1R and subsequent calcium channel were involved in endothelial dysfunction.
Collapse
Affiliation(s)
- Dong-Rui Chen
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Institute of Hypertension, Shanghai, 200025, China
| | - Hui Jiang
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Institute of Hypertension, Shanghai, 200025, China
| | - Jing Chen
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Institute of Hypertension, Shanghai, 200025, China
| | - Cheng-Chao Ruan
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Institute of Hypertension, Shanghai, 200025, China
| | - Wei-Qing Han
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Shanghai Institute of Hypertension, Shanghai, 200025, China.
| | - Ping-Jin Gao
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Institute of Hypertension, Shanghai, 200025, China
| |
Collapse
|
14
|
Li Z, Kong W. Cellular signaling in Abdominal Aortic Aneurysm. Cell Signal 2020; 70:109575. [PMID: 32088371 DOI: 10.1016/j.cellsig.2020.109575] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/19/2020] [Accepted: 02/19/2020] [Indexed: 12/31/2022]
Abstract
Abdominal aortic aneurysms (AAAs) are highly lethal cardiovascular diseases without effective medications. However, the molecular and signaling mechanisms remain unclear. A series of pathological cellular processes have been shown to contribute to AAA formation, including vascular extracellular matrix remodeling, inflammatory and immune responses, oxidative stress, and dysfunction of vascular smooth muscle cells. Each cellular process involves complex cellular signaling, such as NF-κB, MAPK, TGFβ, Notch and inflammasome signaling. In this review, we discuss how cellular signaling networks function in various cellular processes during the pathogenesis and progression of AAA. Understanding the interaction of cellular signaling networks with AAA pathogenesis as well as the crosstalk of different signaling pathways is essential for the development of novel therapeutic approaches to and personalized treatments of AAA diseases.
Collapse
Affiliation(s)
- Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| |
Collapse
|
15
|
Jaminon A, Reesink K, Kroon A, Schurgers L. The Role of Vascular Smooth Muscle Cells in Arterial Remodeling: Focus on Calcification-Related Processes. Int J Mol Sci 2019; 20:E5694. [PMID: 31739395 PMCID: PMC6888164 DOI: 10.3390/ijms20225694] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 12/22/2022] Open
Abstract
Arterial remodeling refers to the structural and functional changes of the vessel wall that occur in response to disease, injury, or aging. Vascular smooth muscle cells (VSMC) play a pivotal role in regulating the remodeling processes of the vessel wall. Phenotypic switching of VSMC involves oxidative stress-induced extracellular vesicle release, driving calcification processes. The VSMC phenotype is relevant to plaque initiation, development and stability, whereas, in the media, the VSMC phenotype is important in maintaining tissue elasticity, wall stress homeostasis and vessel stiffness. Clinically, assessment of arterial remodeling is a challenge; particularly distinguishing intimal and medial involvement, and their contributions to vessel wall remodeling. The limitations pertain to imaging resolution and sensitivity, so methodological development is focused on improving those. Moreover, the integration of data across the microscopic (i.e., cell-tissue) and macroscopic (i.e., vessel-system) scale for correct interpretation is innately challenging, because of the multiple biophysical and biochemical factors involved. In the present review, we describe the arterial remodeling processes that govern arterial stiffening, atherosclerosis and calcification, with a particular focus on VSMC phenotypic switching. Additionally, we review clinically applicable methodologies to assess arterial remodeling and the latest developments in these, seeking to unravel the ubiquitous corroborator of vascular pathology that calcification appears to be.
Collapse
Affiliation(s)
- Armand Jaminon
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Koen Reesink
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Abraham Kroon
- Department of Internal Medicine, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands;
| | - Leon Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| |
Collapse
|
16
|
Wang Z, Guo J, Han X, Xue M, Wang W, Mi L, Sheng Y, Ma C, Wu J, Wu X. Metformin represses the pathophysiology of AAA by suppressing the activation of PI3K/AKT/mTOR/autophagy pathway in ApoE -/- mice. Cell Biosci 2019; 9:68. [PMID: 31467666 PMCID: PMC6712653 DOI: 10.1186/s13578-019-0332-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/16/2019] [Indexed: 02/07/2023] Open
Abstract
Background The protective effect of metformin (MET) on abdominal aortic aneurysm (AAA) has been reported. However, the related mechanism is still poor understood. In this study, we deeply investigated the role of metformin in AAA pathophysiology. Methods Angiotensin II (Ang-II) was used to construct the AAA model in ApoE−/− mice. The related mechanism was explored using Western blot and quantitative real time PCR (qRT-PCR). We also observed the morphological changes in the abdominal aorta and the influence of metformin on biological behaviors of rat abdominal aortic VSMCs. Results The PI3K/AKT/mTOR pathway was activated in aneurysmal wall tissues of AAA patients and rat model. Treatment with metformin inhibited the breakage and preserved the elastin structure of the aorta, the loss of collagen, and the apoptosis of aortic cells. In addition, metformin significantly suppressed the activation of the PI3K/AKT/mToR pathway and decreased the mRNA and protein levels of LC3B and Beclin1, which were induced by Ang-II. Moreover, PI3K inhibitors enhanced the effect of metformin while PI3K agonists largely reversed this effect. Interestingly, the cell proliferation, apoptosis, migration and autophagy of vascular smooth muscle cells (VSMCs) induced by Ang-II were also decreased following metformin treatment. PI3K inhibitors and agonists strengthened and weakened the effects of metformin in VSMCs, respectively. Conclusions Metformin represses the pathophysiology of AAA by inhibiting the activation of PI3K/AKT/mTOR/autophagy pathway. This repression may be useful as a new therapeutic strategy for AAA.
Collapse
Affiliation(s)
- Zhu Wang
- 1Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jing Wu Wei Qi Road, Jinan, 250021 Shandong China.,2Department of Interventional Medicine and Vascular Surgery, Binzhou Medical University Hospital, Binzhou, 256603 Shandong China
| | - Jingjing Guo
- 3Department of Obstetrics and Gynecology, Binzhou Medical University Hospital, Binzhou, 256603 Shandong China
| | - Xinqiang Han
- 2Department of Interventional Medicine and Vascular Surgery, Binzhou Medical University Hospital, Binzhou, 256603 Shandong China
| | - Ming Xue
- 4Department of Interventional Radiology, Weihai Municipal Hospital, Weihai, 264200 Shandong China
| | - Wenming Wang
- 2Department of Interventional Medicine and Vascular Surgery, Binzhou Medical University Hospital, Binzhou, 256603 Shandong China
| | - Lei Mi
- Department of General Surgery, Taian City Central Hospital, Taian, 271000 Shandong China
| | - Yuguo Sheng
- 2Department of Interventional Medicine and Vascular Surgery, Binzhou Medical University Hospital, Binzhou, 256603 Shandong China
| | - Chao Ma
- 2Department of Interventional Medicine and Vascular Surgery, Binzhou Medical University Hospital, Binzhou, 256603 Shandong China
| | - Jian Wu
- 2Department of Interventional Medicine and Vascular Surgery, Binzhou Medical University Hospital, Binzhou, 256603 Shandong China
| | - Xuejun Wu
- 1Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jing Wu Wei Qi Road, Jinan, 250021 Shandong China
| |
Collapse
|
17
|
Plant-Derived Products for Treatment of Vascular Intima Hyperplasia Selectively Inhibit Vascular Smooth Muscle Cell Functions. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:3549312. [PMID: 30405738 PMCID: PMC6201497 DOI: 10.1155/2018/3549312] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 09/01/2018] [Accepted: 09/20/2018] [Indexed: 12/11/2022]
Abstract
Natural products are used widely for preventing intimal hyperplasia (IH), a common cardiovascular disease. Four different cells initiate and progress IH, namely, vascular smooth muscle, adventitial and endothelial cells, and circulation or bone marrow-derived cells. Vascular smooth muscle cells (VSMCs) play a critical role in initiation and development of intimal thickening and formation of neointimal hyperplasia. In this review, we describe the different originating cells involved in vascular IH and emphasize the effect of different natural products on inhibiting abnormal cellular functions, such as VSMC proliferation and migration. We further present a classification for the different natural products like phenols, flavonoids, terpenes, and alkaloids that suppress VSMC growth. Abnormal VSMC physiology involves disturbance in MAPKs, PI3K/AKT, JAK-STAT, FAK, and NF-κB signal pathways. Most of the natural isolate studies have revealed G1/S phase of cell cycle arrest, decreased ROS production, induced cell apoptosis, restrained migration, and downregulated collagen deposition. It is necessary to screen optimal drugs from natural sources that preferentially inhibit VSMC rather than vascular endothelial cell growth to prevent early IH, restenosis following graft implantation, and atherosclerotic diseases.
Collapse
|