1
|
Hytönen JP, Leppänen O, Taavitsainen J, Ylä-Herttuala S. Synthetic Flavonoid 3,7-Dihydroxy-Isoflav-3-Ene (DHIF) Reduces In-Stent Restenosis in an Atherosclerotic Watanabe Heritable Hyperlipidemic Rabbit Stent Model. Int J Mol Sci 2024; 25:11530. [PMID: 39519083 PMCID: PMC11546789 DOI: 10.3390/ijms252111530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Inflammation is a major component of the pathogenesis of atherosclerosis and the formation of in-stent restenosis (ISR). A novel flavonoid, DHIF, attenuates reactive oxygen species and nf-κB signaling and has potential to limit ISR via antioxidant action. While current drug eluting stents (DESs) perform well in clinical practice, new therapies to prevent ISR without dependance on cytotoxic drugs are warranted. Our objective was to test whether DHIF reduces ISR in a hyperlipidemic rabbit aorta model of ISR via attenuated inflammatory responses. WHHL rabbit aortas (n = 24) were denuded. Six weeks after injury, stents were implanted into the denuded aortas. DHIF was dissolved in carboxymethyl cellulose (CMC) and administered orally with two doses. CMC served as a control. The animals were sacrificed six weeks after stenting. ISR was evaluated from stent histomorphometry and immunohistology was used to assess the inflammatory and antiproliferative effects of the treatment. ISR was reduced from 20.9 ± 3.0% in controls to 15.2 ± 2.4% (p = 0.0009) and 16.4 ± 2.1% (p = 0.004) in the low- and high-dose groups, respectively. The neointimal area covered by macrophages was 32 ± 9.3% in the controls, 17.2 ± 5.9% (p = 0.005) in the low-dose group and 19.4 ± 7.9% (p = 0.008) in the high-dose group. DHIF significantly reduces ISR and local inflammation in stented arterial regions and could be used to reduce ISR when bare metal stents are used. Targeting local inflammation in the arterial wall may provide a way to reduce ISR in a clinical setting and further studies are warranted.
Collapse
Affiliation(s)
- Jarkko P. Hytönen
- A.I. Virtanen Institute, University of Eastern Finland, 70210 Kuopio, Finland; (J.P.H.)
- Heart Center, Kuopio University Hospital, 70200 Kuopio, Finland
| | - Olli Leppänen
- A.I. Virtanen Institute, University of Eastern Finland, 70210 Kuopio, Finland; (J.P.H.)
| | - Jouni Taavitsainen
- A.I. Virtanen Institute, University of Eastern Finland, 70210 Kuopio, Finland; (J.P.H.)
- Heart Center, Kuopio University Hospital, 70200 Kuopio, Finland
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute, University of Eastern Finland, 70210 Kuopio, Finland; (J.P.H.)
- Heart Center, Kuopio University Hospital, 70200 Kuopio, Finland
| |
Collapse
|
2
|
Luo Y, Zhang Z, Zheng W, Zeng Z, Fan L, Zhao Y, Huang Y, Cao S, Yu S, Shen L. Molecular Mechanisms of Plant Extracts in Protecting Aging Blood Vessels. Nutrients 2024; 16:2357. [PMID: 39064801 PMCID: PMC11279783 DOI: 10.3390/nu16142357] [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: 06/13/2024] [Revised: 07/09/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Plant Extracts (PE) are natural substances extracted from plants, rich in various bioactive components. Exploring the molecular mechanisms and interactions involved in the vascular protective effects of PE is beneficial for the development of further strategies to protect aging blood vessels. For this review, the content was obtained from scientific databases such as PubMed, China National Knowledge Infrastructure (CNKI), and Google Scholar up to July 2024, using the search terms "Plant extracts", "oxidative stress", "vascular aging", "endothelial dysfunction", "ROS", and "inflammation". This review highlighted the effects of PE in protecting aging blood vessels. Through pathways such as scavenging reactive oxygen species, activating antioxidant signaling pathways, enhancing respiratory chain complex activity, inhibiting mitochondrial-reactive oxygen species generation, improving nitric oxide bioavailability, downregulating the secretion of inflammatory factors, and activating sirtuins 1 and Nrf2 signaling pathways, it can improve vascular structural and functional changes caused by age-related oxidative stress, mitochondrial dysfunction, and inflammation due to aging, thereby reducing the incidence of age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Yuxin Luo
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| | - Zeru Zhang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| | - Weijian Zheng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| | - Zhi Zeng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| | - Lei Fan
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Yuquan Zhao
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| | - Yixin Huang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| | - Suizhong Cao
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| | - Shumin Yu
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| | - Liuhong Shen
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Teaching Animal Hospital, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Z.Z.); (W.Z.); (Z.Z.); (L.F.); (Y.Z.); (Y.H.); (S.C.); (S.Y.)
| |
Collapse
|
3
|
Zong Q, He C, Long B, Huang Q, Chen Y, Li Y, Dang Y, Cai C. Targeted Delivery of Nanoparticles to Blood Vessels for the Treatment of Atherosclerosis. Biomedicines 2024; 12:1504. [PMID: 39062077 PMCID: PMC11275173 DOI: 10.3390/biomedicines12071504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/21/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Atherosclerosis is a common form of cardiovascular disease, which is one of the most prevalent causes of death worldwide, particularly among older individuals. Surgery is the mainstay of treatment for severe stenotic lesions, though the rate of restenosis remains relatively high. Current medication therapy for atherosclerosis has limited efficacy in reversing the formation of atherosclerotic plaques. The search for new drug treatment options is imminent. Some potent medications have shown surprising therapeutic benefits in inhibiting inflammation and endothelial proliferation in plaques. Unfortunately, their use is restricted due to notable dose-dependent systemic side effects or degradation. Nevertheless, with advances in nanotechnology, an increasing number of nano-related medical applications are emerging, such as nano-drug delivery, nano-imaging, nanorobots, and so forth, which allow for restrictions on the use of novel atherosclerotic drugs to be lifted. This paper reviews new perspectives on the targeted delivery of nanoparticles to blood vessels for the treatment of atherosclerosis in both systemic and local drug delivery. In systemic drug delivery, nanoparticles inhibit drug degradation and reduce systemic toxicity through passive and active pathways. To further enhance the precise release of drugs, the localized delivery of nanoparticles can also be accomplished through blood vessel wall injection or using endovascular interventional devices coated with nanoparticles. Overall, nanotechnology holds boundless potential for the diagnosis and treatment of atherosclerotic diseases in the future.
Collapse
Affiliation(s)
- Qiushuo Zong
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Chengyi He
- Department of Vascular Surgery, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China;
| | - Binbin Long
- Department of General Surgery, Taihe Hospital Affiliated to Hubei University of Medicine, Shiyan 442099, China;
| | - Qingyun Huang
- Department of Cardiothoracic Surgery, The First Hospital of Putian Affiliated to Fujian Medical University, Putian 351106, China;
| | - Yunfei Chen
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Yiping Dang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Chuanqi Cai
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| |
Collapse
|
4
|
Jin X, Yue X, Huang Z, Meng X, Xu S, Wu Y, Wan Y, Inoue A, Narisawa M, Hu L, Shi GP, Umegaki H, Murohara T, Lei Y, Kuzuya M, Cheng XW. Cathepsin K deficiency prevented stress-related thrombosis in a mouse FeCl 3 model. Cell Mol Life Sci 2024; 81:205. [PMID: 38703204 PMCID: PMC11069486 DOI: 10.1007/s00018-024-05240-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND Exposure to chronic psychological stress (CPS) is a risk factor for thrombotic cardiocerebrovascular diseases (CCVDs). The expression and activity of the cysteine cathepsin K (CTSK) are upregulated in stressed cardiovascular tissues, and we investigated whether CTSK is involved in chronic stress-related thrombosis, focusing on stress serum-induced endothelial apoptosis. METHODS AND RESULTS Eight-week-old wild-type male mice (CTSK+/+) randomly divided to non-stress and 3-week restraint stress groups received a left carotid artery iron chloride3 (FeCl3)-induced thrombosis injury for biological and morphological evaluations at specific timepoints. On day 21 post-stress/injury, the stress had enhanced the arterial thrombi weights and lengths, in addition to harmful alterations of plasma ADAMTS13, von Willebrand factor, and plasminogen activation inhibitor-1, plus injured-artery endothelial loss and CTSK protein/mRNA expression. The stressed CTSK+/+ mice had increased levels of injured arterial cleaved Notch1, Hes1, cleaved caspase8, matrix metalloproteinase-9/-2, angiotensin type 1 receptor, galactin3, p16IN4A, p22phox, gp91phox, intracellular adhesion molecule-1, TNF-α, MCP-1, and TLR-4 proteins and/or genes. Pharmacological and genetic inhibitions of CTSK ameliorated the stress-induced thrombus formation and the observed molecular and morphological changes. In cultured HUVECs, CTSK overexpression and silencing respectively increased and mitigated stressed-serum- and H2O2-induced apoptosis associated with apoptosis-related protein changes. Recombinant human CTSK degraded γ-secretase substrate in a dose-dependent manor and activated Notch1 and Hes1 expression upregulation. CONCLUSIONS CTSK appeared to contribute to stress-related thrombosis in mice subjected to FeCl3 stress, possibly via the modulation of vascular inflammation, oxidative production and apoptosis, suggesting that CTSK could be an effective therapeutic target for CPS-related thrombotic events in patients with CCVDs.
Collapse
Affiliation(s)
- Xueying Jin
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Xueling Yue
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China.
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan.
| | - Zhe Huang
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
- Department of Neurology, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Xiangkun Meng
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Shengnan Xu
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Yuna Wu
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China
| | - Ying Wan
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Aiko Inoue
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University Institute of Innovation for Future Society, Nagoya University, Nagoya, Aichi-Ken, 466-8550, Japan
| | - Megumi Narisawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Lina Hu
- Department of Public Health, Guilin Medical College, Guilin, 541199, Guangxi, People's Republic of China
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Hiroyuki Umegaki
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University Institute of Innovation for Future Society, Nagoya University, Nagoya, Aichi-Ken, 466-8550, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Yanna Lei
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China.
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan.
- Department of Intensive Care, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China.
| | - Masafumi Kuzuya
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
- Meitetsu Hospital, Nagoya, Aichi, 451-8511, Japan
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China.
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan.
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, 133002, Jilin, People's Republic of China.
| |
Collapse
|
5
|
Teng L, Qin Q, Zhou ZY, Zhou F, Cao CY, He C, Ding JW, Yang J. Role of C/EBP Homologous Protein in Vascular Stenosis After Carotid Artery Injury. Biochem Genet 2024:10.1007/s10528-024-10713-9. [PMID: 38526708 DOI: 10.1007/s10528-024-10713-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/22/2024] [Indexed: 03/27/2024]
Abstract
The study aims to explore the fluctuating expression of C/EBP Homologous Protein (CHOP) following rat carotid artery injury and its central role in vascular stenosis. Using in vivo rat carotid artery injury models and in vitro ischemia and hypoxia cell models employing human aortic endothelial cells (HAECs) and vascular smooth muscle cells (T/G HA-VSMCs), a comprehensive investigative framework was established. Histological analysis confirmed intimal hyperplasia in rat models. CHOP expression in vascular tissues was assessed using Western blot and immunohistochemical staining, and its presence in HAECs and T/G HA-VSMCs was determined through RT-PCR and Western blot. The study evaluated HAEC apoptosis, inflammatory cytokine secretion, cell proliferation, and T/G HA-VSMCs migration through Western blot, ELISA, CCK8, and Transwell migration assays. The rat carotid artery injury model revealed substantial fibrous plaque formation and vascular stenosis, resulting in an increased intimal area and plaque-to-lumen area ratio. Notably, CHOP is markedly elevated in vessels of the carotid artery injury model compared to normal vessels. Atorvastatin effectively mitigated vascular stenosis and suppresses CHOP protein expression. In HAECs, ischemia and hypoxia-induced CHOP upregulation, along with heightened TNFα, IL-6, caspase3, and caspase8 levels, while reducing cell proliferation. Atorvastatin demonstrated a dose-dependent suppression of CHOP expression in HAECs. Downregulation of CHOP or atorvastatin treatment led to reduced IL-6 and TNFα secretion, coupled with augmented cell proliferation. Similarly, ischemia and hypoxia conditions increased CHOP expression in T/G HA-VSMCs, which was concentration-dependently inhibited by atorvastatin. Furthermore, significantly increased MMP-9 and MMP-2 concentrations in the cell culture supernatant correlated with enhanced T/G HA-VSMCs migration. However, interventions targeting CHOP downregulation and atorvastatin usage curtailed MMP-9 and MMP-2 secretion and suppressed cell migration. In conclusion, CHOP plays a crucial role in endothelial injury, proliferation, and VSMCs migration during carotid artery injury, serving as a pivotal regulator in post-injury fibrous plaque formation and vascular remodeling. Statins emerge as protectors of endothelial cells, restraining VSMCs migration by modulating CHOP expression.
Collapse
Affiliation(s)
- Lin Teng
- Department of Cardiology, Yichang Central People's Hospital, NO, 183 Yiling Road, Yichang, 443003, Hubei, People's Republic of China
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, SE5 9NU, UK
| | - Qin Qin
- Department of Cardiology, Yichang Central People's Hospital, NO, 183 Yiling Road, Yichang, 443003, Hubei, People's Republic of China
- School of Basic Medicine, China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Zi-Yi Zhou
- Department of Cardiology, Yichang Central People's Hospital, NO, 183 Yiling Road, Yichang, 443003, Hubei, People's Republic of China
- School of Basic Medicine, China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Fei Zhou
- Department of Cardiology, Yichang Central People's Hospital, NO, 183 Yiling Road, Yichang, 443003, Hubei, People's Republic of China
| | - Cun-Yu Cao
- School of Basic Medicine, China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
- Hubei Key Laboratory of Tumor Microencironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Chao He
- Department of Cardiology, Yichang Central People's Hospital, NO, 183 Yiling Road, Yichang, 443003, Hubei, People's Republic of China
| | - Jia-Wang Ding
- Department of Cardiology, Yichang Central People's Hospital, NO, 183 Yiling Road, Yichang, 443003, Hubei, People's Republic of China
| | - Jian Yang
- Department of Cardiology, Yichang Central People's Hospital, NO, 183 Yiling Road, Yichang, 443003, Hubei, People's Republic of China.
| |
Collapse
|
6
|
Liu W, Ding K, Bao J, Hu Y, Gui Y, Ye L, Wang L. Relationship between uric acid to albumin ratio and in-stent restenosis in patients with coronary artery disease undergoing drug-eluting stenting. Coron Artery Dis 2023; 34:589-594. [PMID: 37855441 PMCID: PMC10602219 DOI: 10.1097/mca.0000000000001300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 09/20/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND In-stent restenosis (ISR) in patients undergoing percutaneous coronary intervention (PCI) to treat coronary artery disease (CAD) is an urgent issue in clinical practice. Recent studies have highlighted uric acid-albumin ratio (UAR) as a new marker for evaluating inflammation and oxidative stress, capable of predicting cardiovascular ailments. We aimed to examine the correlation between UAR levels and ISR in patients who underwent drug-eluting stent (DES) implantation. METHODS We included 503 patients with CAD who underwent initial DES implantation and angiography during the follow-up period. Based on coronary angiographic findings, the patients were categorized into ISR (n = 73) and non-ISR groups (n = 430). Before angiography, laboratory parameters were measured for all enrolled patients. To ascertain the influential factors linked to ISR, multivariate logistic regression analysis was performed. The predictive capability of UAR in determining ISR was assessed using receiver operating characteristic (ROC) curve analysis. Statistical significance was set at P < 0.05. RESULTS Multivariate logistic regression analysis revealed that diabetes mellitus, stent length, UAR, albumin levels, and C-reactive protein levels independently predicted ISR. ROC curve analysis revealed that UAR had an area under the curve of 0.767 (95% CI: 0.709 - 0.826) for predicting ISR and demonstrated that UAR outperformed the individual predictive abilities of uric acid and albumin for ISR. CONCLUSION UAR was associated with ISR in patients with CAD undergoing PCI with DES implantation. Moreover, ROC curve analysis demonstrated that UAR exhibited superior predictive accuracy for ISR compared with evaluating uric acid and albumin levels separately.
Collapse
Affiliation(s)
- Wenquan Liu
- Jinzhou Medical University, Jinzhou, Liaoning Province, China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Kun Ding
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiaqi Bao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yilan Hu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yang Gui
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lifang Ye
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lihong Wang
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Bai X, Zhang W, Yu T. Integrative bioinformatics analysis identifies APOB as a critical biomarker in coronary in-stent restenosis. Biomark Med 2023; 17:983-998. [PMID: 38223945 DOI: 10.2217/bmm-2023-0507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024] Open
Abstract
Aim: Coronary artery disease (CAD) is a major contributor to the worldwide prevalence of cardiovascular disease. In-stent restenosis (ISR) is a common complication which can lead to stent implantation failure, necessitating repeated intervention and presenting a significant obstacle for CAD management. Methods: To accurately assess and determine the hub genes associated with ISR, CAD databases from the Gene Expression Omnibus were utilized and weighted gene coexpression network analysis was employed to identify key genes in blood samples. Results: APOB was identified as a risk gene for ISR occurrence. Subsequent correlation analysis of APOB demonstrated a positive association with ISR. Clinical validation further confirmed the predictive value of APOB in ISR detection. Conclusion: We have identified APOB as a critical predictive biomarker for ISR in CAD patients.
Collapse
Affiliation(s)
- Xinghua Bai
- Department of Cardiovascular Medicine, The First People's Hospital of Linping District, Hangzhou, 311100, PR China
| | - Weizong Zhang
- Department of Cardiovascular Medicine, The First People's Hospital of Linping District, Hangzhou, 311100, PR China
| | - Tao Yu
- Department of Cardiovascular Medicine, The First People's Hospital of Linping District, Hangzhou, 311100, PR China
| |
Collapse
|
8
|
Watanabe Y, Mitomo S, Naganuma T, Nakajima A, Matsuoka S, Tahara S, Okutsu M, Nakamura S, Nakamura S. Impact of Stent Expansion Index on Stent Failure After Left Main Stenting. Am J Cardiol 2023; 205:164-172. [PMID: 37598602 DOI: 10.1016/j.amjcard.2023.07.157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/20/2023] [Accepted: 07/26/2023] [Indexed: 08/22/2023]
Abstract
Impact of the stent expansion index (EXPI) in percutaneous coronary intervention (PCI) for unprotected left main distal bifurcation lesions (ULMD) has been not completely understood especially in current-generation drug-eluting stent (cDES) era. We evaluated the impact of EXPI on clinical outcomes after PCI with cDES for ULMD. We identified 342 patients treated with cDES for ULMD and postintervention intravascular ultrasound between January 2010 and December 2019. In this study, the ratio of minimum stent area (MSA) to reference vessel area at the MSA site was adopted to assess the stent expansion. We defined the patients with the first and second tertile as low-intermediate EXPI group and those with the third tertile as high EXPI group and compared the clinical outcomes between both groups. The primary end point was target lesion failure (TLF). TLF was defined as a composite of cardiac death, target lesion revascularization (TLR) ,and myocardial infarction. The MSA was located in the ostium of left anterior descending coronary artery in most cases (318 of 342 patients; 93.0%). There were no significant differences between both groups in the baseline clinical, lesion, and procedural characteristics. The high EXPI group had lower TLF rate than the low-intermediate EXPI group (10.2% vs 19.9%, log-rank p = 0.033). In conclusion, this is the first report that the higher ratio of MSA to reference vessel area at the MSA site, which was defined as stent EXPI, was associated with more favorable clinical outcomes after PCI for ULMD.
Collapse
Affiliation(s)
- Yusuke Watanabe
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan.
| | - Satoru Mitomo
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Toru Naganuma
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Akihiro Nakajima
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Satoshi Matsuoka
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Satoko Tahara
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Masaaki Okutsu
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Shotaro Nakamura
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Sunao Nakamura
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| |
Collapse
|
9
|
Caiati C, Stanca A, Lepera ME. Free Radicals and Obesity-Related Chronic Inflammation Contrasted by Antioxidants: A New Perspective in Coronary Artery Disease. Metabolites 2023; 13:712. [PMID: 37367870 DOI: 10.3390/metabo13060712] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/28/2023] [Accepted: 05/29/2023] [Indexed: 06/28/2023] Open
Abstract
We are surrounded by factors called free radicals (FR), which attach to the molecules our body is made of, first among them the endothelium. Even though FR are to a certain extent a normal factor, nowadays we face an escalating increase in these biologically aggressive molecules. The escalating formation of FR is linked to the increased usage of man-made chemicals for personal care (toothpaste, shampoo, bubble bath, etc.), domestic laundry and dish-washer detergents, and also an ever wider usage of drugs (both prescription and over the counter), especially if they are to be used long-term (years). In addition, tobacco smoking, processed foods, pesticides, various chronic infectious microbes, nutritional deficiencies, lack of sun exposure, and, finally, with a markedly increasing impact, electromagnetic pollution (a terribly destructive factor), can increase the risk of cancer, as well as endothelial dysfunction, owing to the increased production of FR that they cause. All these factors create endothelial damage, but the organism may be able to repair such damage thanks to the intervention of the immune system supported by antioxidants. However, one other factor can perpetuate the state of inflammation, namely obesity and metabolic syndrome with associated hyperinsulinemia. In this review, the role of FR, with a special emphasis on their origin, and of antioxidants, is explored from the perspective of their role in causing atherosclerosis, in particular at the coronary level.
Collapse
Affiliation(s)
- Carlo Caiati
- Unit of Cardiovascular Diseases, Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Alessandro Stanca
- Unit of Cardiovascular Diseases, Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Mario Erminio Lepera
- Unit of Cardiovascular Diseases, Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| |
Collapse
|
10
|
Liu W, Wang X, Feng Y. Restoring endothelial function: shedding light on cardiovascular stent development. Biomater Sci 2023. [PMID: 37161519 DOI: 10.1039/d3bm00390f] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Complete endothelialization is highly important for maintaining long-term patency and avoiding subsequent complications in implanting cardiovascular stents. It not only refers to endothelial cells (ECs) fully covering the inserted stents, but also includes the newly formed endothelium, which could exert physiological functions, such as anti-thrombosis and anti-stenosis. Clinical outcomes have indicated that endothelial dysfunction, especially the insufficiency of antithrombotic and barrier functions, is responsible for stent failure. Learning from vascular pathophysiology, endothelial dysfunction on stents is closely linked to the microenvironment of ECs. Evidence points to inflammatory responses, oxidative stress, altered hemodynamic shear stress, and impaired endothelial barrier affecting the normal growth of ECs, which are the four major causes of endothelial dysfunction. The related molecular mechanisms and efforts dedicated to improving the endothelial function are emphasized in this review. From the perspective of endothelial function, the design principles, advantages, and disadvantages behind current stents are introduced to enlighten the development of new-generation stents, aiming to offer new alternatives for restoring endothelial function.
Collapse
Affiliation(s)
- Wen Liu
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, P. R. China.
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin 300072, P. R. China
| | - Xiaoyu Wang
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, P. R. China.
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin 300072, P. R. China
| | - Yakai Feng
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, P. R. China.
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin 300072, P. R. China
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Weijin Road 92, Tianjin 300072, P. R. China
- Frontiers Science Center for Synthetic Biology, Tianjin University, Weijin Road 92, Tianjin 300072, China
| |
Collapse
|
11
|
Talebian S, Mendes B, Conniot J, Farajikhah S, Dehghani F, Li Z, Bitoque D, Silva G, Naficy S, Conde J, Wallace GG. Biopolymeric Coatings for Local Release of Therapeutics from Biomedical Implants. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207603. [PMID: 36782094 PMCID: PMC10131825 DOI: 10.1002/advs.202207603] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Indexed: 06/18/2023]
Abstract
The deployment of structures that enable localized release of bioactive molecules can result in more efficacious treatment of disease and better integration of implantable bionic devices. The strategic design of a biopolymeric coating can be used to engineer the optimal release profile depending on the task at hand. As illustrative examples, here advances in delivery of drugs from bone, brain, ocular, and cardiovascular implants are reviewed. These areas are focused to highlight that both hard and soft tissue implants can benefit from controlled localized delivery. The composition of biopolymers used to achieve appropriate delivery to the selected tissue types, and their corresponding outcomes are brought to the fore. To conclude, key factors in designing drug-loaded biopolymeric coatings for biomedical implants are highlighted.
Collapse
Affiliation(s)
- Sepehr Talebian
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- Nano Institute (Sydney Nano)The University of SydneySydneyNSW2006Australia
| | - Bárbara Mendes
- ToxOmicsNOVA Medical School|Faculdade de Ciências MédicasNMS|FCMUniversidade Nova de LisboaLisboa1169‐056Portugal
| | - João Conniot
- ToxOmicsNOVA Medical School|Faculdade de Ciências MédicasNMS|FCMUniversidade Nova de LisboaLisboa1169‐056Portugal
| | - Syamak Farajikhah
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- Nano Institute (Sydney Nano)The University of SydneySydneyNSW2006Australia
| | - Fariba Dehghani
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- Nano Institute (Sydney Nano)The University of SydneySydneyNSW2006Australia
| | - Zhongyan Li
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
| | - Diogo Bitoque
- ToxOmicsNOVA Medical School|Faculdade de Ciências MédicasNMS|FCMUniversidade Nova de LisboaLisboa1169‐056Portugal
| | - Gabriela Silva
- ToxOmicsNOVA Medical School|Faculdade de Ciências MédicasNMS|FCMUniversidade Nova de LisboaLisboa1169‐056Portugal
| | - Sina Naficy
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- Nano Institute (Sydney Nano)The University of SydneySydneyNSW2006Australia
| | - João Conde
- ToxOmicsNOVA Medical School|Faculdade de Ciências MédicasNMS|FCMUniversidade Nova de LisboaLisboa1169‐056Portugal
| | - Gordon G. Wallace
- Intelligent Polymer Research InstituteARC Centre of Excellence for Electromaterials ScienceAIIM FacilityUniversity of WollongongSydneyNSW2522Australia
| |
Collapse
|
12
|
Liu L, Lan X, Chen X, Dai S, Wang Z, Zhao A, Lu L, Huang N, Chen J, Yang P, Liao Y. Multi-functional plant flavonoids regulate pathological microenvironments for vascular stent surface engineering. Acta Biomater 2023; 157:655-669. [PMID: 36436757 DOI: 10.1016/j.actbio.2022.11.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 10/23/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Abstract
In-stent restenosis (ISR) and late thrombosis, usually caused by excessive smooth muscle cell (SMC) proliferation and delayed endothelial layer repair, respectively, are the main risks for the failure of vascular stent implantation. For years, modification of stents with biomolecules that could selectively inhibit SMC proliferation and support endothelial cell (EC) growth had drawn extensive attention. However, the modulatory effect of these biomolecules faces the impact of oxidative stress, inflammation, and hyperlipidemia of the pathological vascular microenvironment, which is caused by the stent implantation injury and atherosclerosis lesions. Here, we modified stents with a natural and multi-functional flavonoid, baicalin (BCL), using poly-dopamine (PDA) coating technology to combat the harmful impact of the pathological microenvironment. Stent with an appropriate BCL immobilization density (approximately 2.03 μg/cm2) successfully supported ECs growth while inhibited SMC proliferation. Furthermore, baicalin-modified surfaces regulated the oxidative stress, inflammation, and high-lipid of the pathological microenvironment to inhibit endothelial dysfunction and the oxidized low-density lipoprotein (ox-LDL)-induced macrophage foam cells formation. In vivo results showed that baicalin-modified stents exhibited significant anti-ISR, anti-inflammatory, and endothelialization-promoting functions. Our study suggests that the multi-functional baicalin with pathological microenvironment-regulation (PMR) effect has potential use in the surface engineering of cardiovascular devices. STATEMENT OF SIGNIFICANCE: Empowering vascular stents with selective modulation of smooth muscle cells and endothelial cells by surface technology has become an important research direction for stent surface engineering. However, stent coatings that can furthermodulate the pathological microenvironment of blood vessels have been rarely reported. In this study, we constructed a multifunctional coating based on a flavonoid, baicalin, which can selectively modulate vascular wall cells and improve the pathological microenvironment. This study may provide a reference for developing advanced vascular stents.
Collapse
Affiliation(s)
- Luying Liu
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Xiaorong Lan
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou 646000, China
| | - Xiao Chen
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Sheng Dai
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Zhixing Wang
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Ansha Zhao
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Lei Lu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Nan Huang
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Jiang Chen
- Sichuan Key Laboratory for Disease Gene Study, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32 of the West Second Section of First Ring Road, Chengdu 610031, PR China.
| | - Ping Yang
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China.
| | - Yuzhen Liao
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China.
| |
Collapse
|
13
|
Wang R, Lu J, Yin J, Chen H, Liu H, Xu F, Zang T, Xu R, Li C, Wu Y, Wu Q, Fei X, Zhu M, Shen L, Ge J. A TEMPOL and rapamycin loaded nanofiber-covered stent favors endothelialization and mitigates neointimal hyperplasia and local inflammation. Bioact Mater 2023; 19:666-677. [PMID: 35600979 PMCID: PMC9114161 DOI: 10.1016/j.bioactmat.2022.04.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/12/2022] [Accepted: 04/28/2022] [Indexed: 10/26/2022] Open
|
14
|
Wu T, Li N, Zhang Q, Liu R, Zhao H, Fan Z, Zhuo L, Yang Y, Xu Y. MKL1 fuels ROS-induced proliferation of vascular smooth muscle cells by modulating FOXM1 transcription. Redox Biol 2022; 59:102586. [PMID: 36587486 PMCID: PMC9823229 DOI: 10.1016/j.redox.2022.102586] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
Reactive oxygen species (ROS) promotes vascular injury and neointima formation in part by stimulating proliferation of vascular smooth muscle cells (VSMC). The underlying transcriptional mechanism, however, is not completely understood. Here we report that VSMC-specific deletion of MKL1 in mice suppressed neointima formation in a classic model of vascular injury. Likewise, pharmaceutical inhibition of MKL1 activity by CCG-1423 similarly mollified neointima formation in mice. Over-expression of a constitutively active MKL1 in vascular smooth muscle cells enhanced proliferation in a ROS-dependent manner. On the contrary, MKL1 depletion or inhibition attenuated VSMC proliferation. PCR array based screening identified forkhead box protein M1 (FOXM1) as a direct target for MKL1. MKL1 interacted with E2F1 to activate FOXM1 expression. Concordantly, FOXM1 depletion ameliorated MKL1-dependent VSMC proliferation. Of interest, ROS-induced MKL1 phosphorylation through MK2 was essential for its interaction with E2F1 and consequently FOXM1 trans-activation. Importantly, a positive correlation between FOXM1 expression and VSMC proliferation was identified in arterial specimens from patients with restenosis. Taken together, our data suggest that a redox-sensitive phosphorylation-switch of MKL1 activates FOXM1 transcription and mediates ROS fueled vascular smooth muscle proliferation. Targeting the MK-2/MKL1/FOXM1 axis may be considered as a reasonable approach for treatment of restenosis.
Collapse
Affiliation(s)
- Teng Wu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Nan Li
- Department of Human Anatomy, Nanjing Medical University, Nanjing, China
| | - Qiumei Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Ruiqi Liu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Hongwei Zhao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhiwen Fan
- Department of Pathology, Nanjing Drum Tower Hospital Affiliated with Nanjing University School of Medicine, Nanjing, China
| | - Lili Zhuo
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Yuyu Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China; Institute of Biomedical Research and College of Life Sciences, Liaocheng University, Liaocheng, China.
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China; Institute of Biomedical Research and College of Life Sciences, Liaocheng University, Liaocheng, China.
| |
Collapse
|
15
|
Watanabe Y, Mitomo S, Naganuma T, Takagi K, Kawamoto H, Matsuoka S, Chieffo A, Montorfano M, Nakamura S, Colombo A. The impact of chronic kidney disease severity on clinical outcomes after current generation drug-eluting stent implantation for left main distal bifurcation lesions: the Milan and New-Tokyo registry. SCAND CARDIOVASC J 2022; 56:236-242. [PMID: 35799477 DOI: 10.1080/14017431.2022.2084561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Objectives. The impact of chronic kidney disease (CKD) on clinical outcomes after percutaneous coronary intervention (PCI) for unprotected left main distal bifurcation lesions (ULMD) is not fully understood in current generation drug eluting stent (cDES) era. We assessed clinical outcomes after PCI using cDES for ULMD according to CKD severity based on estimated glomerular filtration rate (eGFR). Design. We identified 720 consecutive patients who underwent PCI using cDES for ULMD at three high volume centers between January 2005 and December 2015. We divided those patients to the following five groups according to eGFR. Each group was defined as follows: no CKD (60 mL/min/1.73 m2 ≤ eGFR), mild CKD (45 ≤ eGFR < 60 mL/min/1.73 m2), moderate CKD (30 ≤ eGFR < 45 mL/min/1.73 m2), severe CKD (15 ≤ eGFR < 30 mL/min/1.73 m2) and hemodialysis (HD). The primary endpoint was target lesion failure (TLF) at 3 years. TLF was defined as a composite of cardiac death, target lesion revascularization (TLR) and myocardial infarction (MI). Results. TLF occurred more frequently in severe CKD and HD group compared with other three groups. Conclusions. The patients who have severe CKD or are on HD, were extremely associated with worse clinical outcomes after PCI for ULMD even with cDES.
Collapse
Affiliation(s)
- Yusuke Watanabe
- Interventional Cardiology Unit, San Raffaele Scientific Institute, Milan, Italy.,Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Satoru Mitomo
- Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Toru Naganuma
- Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan.,Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kensuke Takagi
- Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Hiroyoshi Kawamoto
- Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan.,Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Matsuoka
- Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Alaide Chieffo
- Interventional Cardiology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Montorfano
- Interventional Cardiology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Sunao Nakamura
- Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Antonio Colombo
- Interventional Cardiology Unit, EMO-GVM, Centro Cuore Columbus, Milan, and Villa Maria Cecilia Hospital GVM, Lugo, Italy
| |
Collapse
|
16
|
Advancements of Prussian blue-based nanoplatforms in biomedical fields: Progress and perspectives. J Control Release 2022; 351:752-778. [DOI: 10.1016/j.jconrel.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/02/2022] [Accepted: 10/03/2022] [Indexed: 12/07/2022]
|
17
|
Zhang H, Zhang W, Qiu H, Zhang G, Li X, Qi H, Guo J, Qian J, Shi X, Gao X, Shi D, Zhang D, Gao R, Ding J. A Biodegradable Metal-Polymer Composite Stent Safe and Effective on Physiological and Serum-Containing Biomimetic Conditions. Adv Healthc Mater 2022; 11:e2201740. [PMID: 36057108 DOI: 10.1002/adhm.202201740] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/25/2022] [Indexed: 01/28/2023]
Abstract
The new-generation coronary stents are expected to be biodegradable, and then the biocompatibility along with biodegradation becomes more challenging. It is a critical issue to choose appropriate biomimetic conditions to evaluate biocompatibility. Compared with other candidates for biodegradable stents, iron-based materials are of high mechanical strength, yet have raised more concerns about biodegradability and biocompatibility. Herein, a metal-polymer composite strategy is applied to accelerate the degradation of iron-based stents in vitro and in a porcine model. Furthermore, it is found that serum, the main environment of vascular stents, ensured the safety of iron corrosion through its antioxidants. This work highlights the importance of serum, particularly albumin, for an in vitro condition mimicking blood-related physiological condition, when reactive oxygen species, inflammatory response, and neointimal hyperplasia are concerned. The resultant metal-polymer composite stent is implanted into a patient in clinical research via interventional treatment, and the follow-up confirms its safety, efficacy, and appropriate biodegradability.
Collapse
Affiliation(s)
- Hongjie Zhang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, P. R. China
| | - Wanqian Zhang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, P. R. China.,National and Local Joint Engineering Laboratory of Interventional Medical Biotechnology and System, Biotyx Medical (Shenzhen) Co., Ltd, Lifetech Scientific (Shenzhen) Co. Ltd., Shenzhen, 518110, P. R. China
| | - Hong Qiu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, P. R. China
| | - Gui Zhang
- National and Local Joint Engineering Laboratory of Interventional Medical Biotechnology and System, Biotyx Medical (Shenzhen) Co., Ltd, Lifetech Scientific (Shenzhen) Co. Ltd., Shenzhen, 518110, P. R. China
| | - Xin Li
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, P. R. China
| | - Haiping Qi
- National and Local Joint Engineering Laboratory of Interventional Medical Biotechnology and System, Biotyx Medical (Shenzhen) Co., Ltd, Lifetech Scientific (Shenzhen) Co. Ltd., Shenzhen, 518110, P. R. China
| | - Jingzhen Guo
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, P. R. China
| | - Jie Qian
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, P. R. China
| | - Xiaoli Shi
- National and Local Joint Engineering Laboratory of Interventional Medical Biotechnology and System, Biotyx Medical (Shenzhen) Co., Ltd, Lifetech Scientific (Shenzhen) Co. Ltd., Shenzhen, 518110, P. R. China
| | - Xian Gao
- National and Local Joint Engineering Laboratory of Interventional Medical Biotechnology and System, Biotyx Medical (Shenzhen) Co., Ltd, Lifetech Scientific (Shenzhen) Co. Ltd., Shenzhen, 518110, P. R. China
| | - Daokun Shi
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, P. R. China
| | - Deyuan Zhang
- National and Local Joint Engineering Laboratory of Interventional Medical Biotechnology and System, Biotyx Medical (Shenzhen) Co., Ltd, Lifetech Scientific (Shenzhen) Co. Ltd., Shenzhen, 518110, P. R. China
| | - Runlin Gao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, P. R. China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, P. R. China
| |
Collapse
|
18
|
Age-Related Changes in Skeletal Muscle Oxygen Utilization. J Funct Morphol Kinesiol 2022; 7:jfmk7040087. [PMID: 36278748 PMCID: PMC9590092 DOI: 10.3390/jfmk7040087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
The cardiovascular and skeletal muscle systems are intrinsically interconnected, sharing the goal of delivering oxygen to metabolically active tissue. Deficiencies within those systems that affect oxygen delivery to working tissues are a hallmark of advancing age. Oxygen delivery and utilization are reflected as muscle oxygen saturation (SmO2) and are assessed using near-infrared resonance spectroscopy (NIRS). SmO2 has been observed to be reduced by ~38% at rest, ~24% during submaximal exercise, and ~59% during maximal exercise with aging (>65 y). Furthermore, aging prolongs restoration of SmO2 back to baseline by >50% after intense exercise. Regulatory factors that contribute to reduced SmO2 with age include blood flow, capillarization, endothelial cells, nitric oxide, and mitochondrial function. These mechanisms are governed by reactive oxygen species (ROS) at the cellular level. However, mishandling of ROS with age ultimately leads to alterations in structure and function of the regulatory factors tasked with maintaining SmO2. The purpose of this review is to provide an update on the current state of the literature regarding age-related effects in SmO2. Furthermore, we attempt to bridge the gap between SmO2 and associated underlying mechanisms affected by aging.
Collapse
|
19
|
Hong Q, Que D, Zhong C, Huang G, Zhai W, Chen D, Yan J, Yang P. Trimethylamine-N-oxide (TMAO) promotes balloon injury-induced neointimal hyperplasia via upregulating Beclin1 and impairing autophagic flux. Biomed Pharmacother 2022; 155:113639. [PMID: 36088853 DOI: 10.1016/j.biopha.2022.113639] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND AIMS TMAO is a microbiota-dependent metabolite associated with increased risk of various cardiovascular diseases. However, the relationship between TMAO and vascular injury-related neointimal hyperplasia is unclear. This study aimed to explore whether TMAO promotes neointimal hyperplasia after balloon injury and elucidate the underlying mechanism. METHODS AND RESULTS Through hematoxylin and eosin staining and immunohistochemistry staining, we found that supplementary TMAO promoted balloon injury-induced neointimal hyperplasia, while reducing TMAO by antibiotic administration produced the opposite result. TMAO showed limited effect on rat aortic vascular smooth muscle cells (RAOSMCs) proliferation and migration. However, TMAO notably induced dysfunction of rat aortic vascular endothelial cells (RAOECs) in vitro and attenuated reendothelialization of carotid arteries after balloon injury in vivo. Autophagic flux was measured by fluorescent mRFP-GFP-LC3, transmission electron microscopy, and western blot. TMAO impaired autophagic flux, as evidenced by the accumulation of p62 and LC3II and high autophagosome to autolysosome ratios. Furthermore, we confirmed that Beclin1 level increased in TMAO-treated RAOECs and carotid arteries. Knocking down Beclin1 alleviated TMAO-induced autophagic flux impairment and neointimal hyperplasia. CONCLUSIONS TMAO promoted neointimal hyperplasia through Beclin1-induced autophagic flux blockage, suggesting that TMAO is a potential target for improvement of vascular remodeling after injury.
Collapse
Affiliation(s)
- Qingqing Hong
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong, China; Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong, China
| | - Dongdong Que
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong, China; Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong, China
| | - Chongbin Zhong
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong, China; Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong, China
| | - Guanlin Huang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong, China; Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong, China
| | - Weicheng Zhai
- Department of Cardiology, Huizhou Third People's Hospital, Guangzhou Medical University, Huizhou City, China
| | - Deshu Chen
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong, China; Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong, China
| | - Jing Yan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong, China; Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong, China.
| | - Pingzhen Yang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong, China; Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong, China.
| |
Collapse
|
20
|
Ganjali S, Mansouri A, Abbasifard M, Moallem SA, Tayarani-Najaran Z, Sahebkar A. Association between Oxidative Burden and Restenosis: A Case-Control Study. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3577761. [PMID: 35799893 PMCID: PMC9256427 DOI: 10.1155/2022/3577761] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/01/2022] [Accepted: 06/16/2022] [Indexed: 11/17/2022]
Abstract
Background In-stent restenosis (ISR) is an important clinical complication that occurs following stent implantation. The application of drug-eluting stents (DES) and even consumption of drugs such as antiplatelet agents and statins are not completely effective in reducing ISR risk. Since the number of these patients continues to rise, it is pivotal to detect patients who are at a higher risk of ISR. In addition, identification of biochemical markers of ISR could give the right perspective on choosing the proper strategy to treat these patients. Several pathophysiological pathways including oxidative stress (OS) are implicated in the progression of ISR. Hence, this study aimed to evaluate the association between oxidative/anti-oxidative markers and ISR. Methods This was a case-control study which comprised 21 ISR, 26 NISR (non-ISR), and 20 healthy subjects. The serum levels of OS markers including malondialdehyde (MDA), thiol groups (GSH), total antioxidant capacity (TAC), and the activity of serum antioxidant enzymes such as glutathione peroxidase (GPx) and superoxide dismutase (SOD) were assessed by colorimetric methods. The overall oxidative burden was assessed using a pro-oxidant-antioxidant balance (PAB) assay. Results MDA levels were considerably higher in the ISR group when compared to healthy subjects (P = 0.004). PAB also indicated significantly higher values in both ISR (P < 0.001) and NISR (P < 0.001) groups related to healthy subjects. No significant differences were observed between the studied groups regarding thiol levels, antioxidant enzyme activities, and TAC. Multinomial logistic regression analysis showed that elevated serum levels of MDA (OR: 1.028, 95% CI: 1.008-1.048; P = 0.006) and PAB (OR: 1.076, 95% CI: 1.017-1.139; P = 0.011) were significantly associated with higher ISR risk; however, increased values of TAC (OR: 0.990, 95% CI: 0.982-0.999; P = 0.030) were significantly associated with decreased ISR risk, while after adjustment for confounders, only SOD activity (OR: 0.0, 95% CI: 0.0-0.0; P < 0.001) and PAB value (OR: 1.866, 95% CI: 1.856-1.900; P < 0.001) showed association with ISR risk. Conclusion According to the present findings, some oxidative and antioxidative markers like PAB and SOD activity showed the potential in the prediction of ISR risk.
Collapse
Affiliation(s)
- Shiva Ganjali
- Department of Medical Biotechnology and Nanotechnology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atena Mansouri
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Cellular & Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Mitra Abbasifard
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Internal Medicine, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Seyed Adel Moallem
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Zahraa University for Women, Karbala, Iraq
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Tayarani-Najaran
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
21
|
Kim C, Lee SG, Lim S, Jung M, Kwon SP, Hong J, Kang M, Sohn HS, Go S, Moon S, Lee SJ, Kim JS, Kim BS. A Senolytic-Eluting Coronary Stent for the Prevention of In-Stent Restenosis. ACS Biomater Sci Eng 2022; 8:1921-1929. [PMID: 35416659 DOI: 10.1021/acsbiomaterials.1c01611] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The vast majority of drug-eluting stents (DES) elute either sirolimus or one of its analogues. While limus drugs stymie vascular smooth muscle cell (VSMC) proliferation to prevent in-stent restenosis, their antiproliferative nature is indiscriminate and limits healing of the endothelium in stented vessels, increasing the risk of late-stent thrombosis. Oxidative stress, which is associated with vascular injury from stent implantation, can induce VSMCs to undergo senescence, and senescent VSMCs can produce pro-inflammatory cytokines capable of inducing proliferation of neighboring nonsenescent VSMCs. We explored the potential of senolytic therapy, which involves the selective elimination of senescent cells, in the form of a senolytic-eluting stent (SES) for interventional cardiology. Oxidative stress was modeled in vitro by exposing VSMCs to H2O2, and H2O2-mediated senescence was evaluated by cytochemical staining of senescence-associated β-galactosidase activity and qRT-PCR. Quiescent VSMCs were then treated with the conditioned medium (CM) of H2O2-treated VSMCs. Proliferative effects of CM were analyzed by staining for proliferating cell nuclear antigen. Senolytic effects of the first-generation senolytic ABT263 were observed in vitro, and the effects of ABT263 on endothelial cells were also investigated through an in vitro re-endothelialization assay. SESs were prepared by dip coating. Iliofemoral arteries of hypercholesteremic rabbits were implanted with SES, everolimus-eluting stents (EESs), or bare-metal stents (BMSs), and the area of stenosis was measured 4 weeks post-implantation using optical coherence tomography. We found that a portion of H2O2-treated VSMCs underwent senescence, and that CM of H2O2-treated senescent VSMCs triggered the proliferation of quiescent VSMCs. ABT263 reverted H2O2-mediated senescence and the proliferative capacity of senescent VSMC CM. Unlike everolimus, ABT263 did not affect endothelial cell migration and/or proliferation. SES, but not EES, significantly reduced stenosis area in vivo compared with bare-metal stents (BMSs). This study shows the potential of SES as an alternative to current forms of DES.
Collapse
Affiliation(s)
- Cheesue Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Seul-Gee Lee
- Yonsei Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Songhyun Lim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Mungyo Jung
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sung Pil Kwon
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Mikyung Kang
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Hee Su Sohn
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Seokhyeong Go
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sangjun Moon
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Seung-Jun Lee
- Cardiology Division, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jung-Sun Kim
- Yonsei Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.,Cardiology Division, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea.,Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea.,Institute of Chemical Processes, Institute of Engineering Research, and BioMAX, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
22
|
ZHANG J, MA Y, WANG Z, LI Y, WANG H, WANG Q. Association between Green Tea Consumption and In-Stent Restenosis in a Chinese Population after Percutaneous Coronary Intervention. J Nutr Sci Vitaminol (Tokyo) 2022; 68:120-126. [DOI: 10.3177/jnsv.68.120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Jing ZHANG
- Department of Ultrasound, Zhongda Hospital, Southeast University
| | - Yao MA
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University
| | - Zemu WANG
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University
| | - Yafei LI
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University
| | - Hao WANG
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University
| | - Qiming WANG
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University
| |
Collapse
|
23
|
Jiang Y, Zhao Q, Li L, Huang S, Yi S, Hu Z. Effect of Traditional Chinese Medicine on the Cardiovascular Diseases. Front Pharmacol 2022; 13:806300. [PMID: 35387325 PMCID: PMC8978630 DOI: 10.3389/fphar.2022.806300] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/31/2022] [Indexed: 02/03/2023] Open
Abstract
Background: Traditional Chinese medicine (TCM) is the health care system developed with the help of clinical trials that are based ideally on the scientific model of regulation. Objective: This systematic health care system relies on some specific unique theories and practical experiences to treat and cure diseases, thus enhancing the public's health. Review Methodology: The current review covers the available literature from 2000 to 2021. The data was collected from journals research articles, published books, thesis, and electronic databases, search engines such as Google Scholar, Elsevier, EBSCO, PMC, PubMed, ScienceDirect, Willey Online Library, Springer Link, and CNKI) searching key terms, cardiovascular disease, traditional Chinese medicines, natural products, and bioactive compounds. Full-length articles and abstracts were screened for the collection of information included in the paper. Results: Clinical trials on the TCM and basic research carried out on its mechanism and nature have led to the application and development of the perfect design of the research techniques, for example, twofold striking in acupuncture that aid in overcoming the limitations and resistances in integrating and applicability of these experiences and trials into the pre-existing biomedical models. Furthermore, TCM has also been utilized from ancient times to treat heart diseases in Asia, particularly in China, and is now used by people in many other areas. Cardiovascular disease (CVD) is mainly developed by oxidative stress. Hence antioxidants can be beneficial in treating this particular disease. TCM has a wide variety of antioxidant components. Conclusion: The current review article summarizes the underlying therapeutic property of TCM and its mechanism. It also overviews the evidence of the mechanism of TCM action in CVD prevention by controlling oxidative stress and its signaling pathway.
Collapse
Affiliation(s)
- Yang Jiang
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China.,Hunan Academy of Traditional Chinese Medicine Affiliated Hospital, Changsha, China
| | - Qi Zhao
- Hunan Academy of Traditional Chinese Medicine Affiliated Hospital, Changsha, China
| | - Lin Li
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Shumin Huang
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Shuai Yi
- Hunan Academy of Traditional Chinese Medicine Affiliated Hospital, Changsha, China
| | - Zhixi Hu
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
24
|
Differential Expression of miRNA-223 in Coronary In-Stent Restenosis. J Clin Med 2022; 11:jcm11030849. [PMID: 35160300 PMCID: PMC8836934 DOI: 10.3390/jcm11030849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/03/2022] [Accepted: 01/27/2022] [Indexed: 12/11/2022] Open
Abstract
Objective: In-stent restenosis (ISR) is an unfavorable complication that occurs in patients after coronary stenting. Despite the progress with advent of modern DES and new antiplatelet agents, restenosis still hampers PCI short- and long-term results. The aim of this study was to investigate whether circulating miRNA-223, which is associated with HDL particles and involved in cholesterol efflux pathway, have diagnostic capability for determining ISR. Methods: This case–control study comprised 21 ISR and 26 NISR patients. The level of miRNA-223 expression was evaluated by TaqMan Real-Time PCR, quantified by the comparative method (fold change) and normalized to U6 expression. Results: Patients in ISR and NISR groups were not different in terms of demographic, clinical, and biochemical parameters, except that the percentage of patients who had DES was significantly greater in the NISR group (88.9%) in comparison with the ISR group (50%). The serum expression of miRNA-223 in ISR patients was 3.277 ± 0.9 times greater than that in NISR group (p = 0.016). In addition, the results of binary logistic regression demonstrated that the high level of serum miRNA-223 was strongly and positively associated with the ISR risk (OR: 17.818, 95% CI: 1.115–284.623, p = 0.042) after adjustment for age, sex, HDL-C, LDL-C, FBS, and statin consumption. Conclusion: Elevated serum level of miRNA-223 might be helpful in predicting the occurrence of ISR. Further confirmation in future large-scale studies is warranted.
Collapse
|
25
|
Sato R, Akita K, Ikoma T, Iguchi K, Murase T, Nakamura T, Akari S, Mogi S, Naruse Y, Ohtani H, Maekawa Y. Association between plasma xanthine oxidoreductase activity and in-hospital outcomes in patients with stable coronary artery disease after percutaneous coronary intervention. PLoS One 2021; 16:e0257227. [PMID: 34520495 PMCID: PMC8439489 DOI: 10.1371/journal.pone.0257227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 08/26/2021] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES Reactive oxygen species generated by xanthine oxidoreductase (XOR) are associated with the progression of atherosclerosis. However, changes in plasma XOR (pXOR) activity after percutaneous coronary intervention (PCI) for stable coronary artery disease (CAD) remains unknown. METHODS Herein, we compared the change in the pXOR activity in patients undergoing PCI with that in patients undergoing coronary angiography (CAG) and further evaluated the relation between changes in pXOR activity and in-hospital and long-term outcomes of patients undergoing PCI. The pXOR activity of 80 consecutive patients who underwent PCI and 25 patients who underwent CAG during the hospitalization was analyzed daily. The percentage changes from baseline regulated time interval was evaluated. RESULTS We found that although pXOR activity decreased after PCI, and remained low until discharge, no significant changes were observed in patients undergoing CAG. Furthermore, among the patients undergoing PCI, those who experienced in-hospital adverse events, had a higher percentage of pXOR reduction 3 days after PCI. There was no association between these changes and long-term events. CONCLUSIONS A significant change in pXOR activity was observed in patients undergoing PCI than in patients undergoing CAG, and there seems to be a correlation between the in-hospital outcomes and the percentage reduction from baseline in pXOR activity.
Collapse
Affiliation(s)
- Ryota Sato
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Keitaro Akita
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takenori Ikoma
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Keisuke Iguchi
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | | | - Seigo Akari
- Sanwa Kagaku Kenkyusho Co., Ltd., Aichi, Japan
| | - Satoshi Mogi
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yoshihisa Naruse
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hayato Ohtani
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuichiro Maekawa
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu, Japan
- * E-mail:
| |
Collapse
|
26
|
Zaghloul MS, Abdul-Malak OM, Cherfan P, Go C, Saadeddin Z, Al-Khoury GE, Chaer RA, Avgerinos ED. Female Gender is a Predictor of Lower Iliac Vein Stenting Patency Rates. Ann Vasc Surg 2021; 78:247-256. [PMID: 34464730 DOI: 10.1016/j.avsg.2021.06.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/29/2021] [Accepted: 06/06/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Iliac venous stenting (IVS) for thrombotic and nonthrombotic venous disease is increasingly used as evidence of the safety, efficacy and durability of these interventions increases. Female gender has been implicated as a predictor of failure in arterial endovascular interventions. We hypothesize that female gender could be predictive of patency rates of iliac vein stenting. METHODS Consecutive patients who underwent IVS for thrombotic or nonthrombotic venous disease at our institution from 2007 until 2019 were identified and divided into groups based on gender. Operative notes, venograms, and the electronic health record were then queried to obtain operative details, co-morbid conditions, postoperative outcomes and stent patency. Study outcome was long term patency rate. The data was analyzed using chi-square, logistic regression, and Kaplan-Meier analysis as appropriate. RESULTS A total of 200 consecutive patients (231 limbs) were identified in our retrospective analysis, with a mean age of 48.8 ± 17.3, and BMI of 31.6 ± 8.6. Of those, 119 (59.5%) patients, (131 [56.8%] limbs) were female. Comparisons between the gender groups revealed no difference in age, BMI, or preoperative comorbidities. There was no difference in type of venous disease between male (85% thrombotic, 15% nonthrombotic) and female (84% thrombotic, 16% nonthrombotic), P= 0.830. The male cohort was more likely to present with leg ulceration (17% vs. 4.6%, P = 0.002), and the female cohort was more likely to present with leg edema (98.5% vs. 93.0%, P= 0.03). The male cohort had a higher rate of caval (48% vs. 33.6%, P= 0.027) and infrainguinal stent extension. (11% vs. 6.9%, P= 0.02). Females had a higher rate of left sided stenting (80.9% vs. 66/0%, P= 0.010). There was no difference in the median stent diameter used between the cohorts. Primary patency at 5 years was significantly higher for the male cohort (94.1% vs. 74.4%, P= 0.01) On adjusted multivariable cox regression female gender was a predictor of loss of primary patency within 5 years (HR, 4.04; P= 0.007). CONCLUSIONS In this single center retrospective analysis of IVS, male patients were found to have better primary stent patency compared to female.
Collapse
Affiliation(s)
- Mohamed S Zaghloul
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Othman M Abdul-Malak
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Patrick Cherfan
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Catherine Go
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Zein Saadeddin
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Georges E Al-Khoury
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Rabih A Chaer
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Efthymios D Avgerinos
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA.
| |
Collapse
|
27
|
Pepe M, Napoli G, Carulli E, Moscarelli M, Forleo C, Nestola PL, Biondi-Zoccai G, Giordano A, Favale S. Autoimmune diseases in patients undergoing percutaneous coronary intervention: A risk factor for in-stent restenosis? Atherosclerosis 2021; 333:24-31. [PMID: 34418682 DOI: 10.1016/j.atherosclerosis.2021.08.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/25/2021] [Accepted: 08/04/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Despite the relation between autoimmune diseases and increased atherosclerotic risk is established, the influence of autoimmune disorders on in-stent restenosis (ISR) after percutaneous coronary intervention (PCI) is only partly known. ISR is an aberrant reparative process mainly characterized by an increased number of vascular smooth muscle cells and excessive deposition of extracellular proteoglycans and type III collagen. Chronic inflammation, always present in autoimmune diseases, modulates the endothelial response to PCI. Aim of this review is to resume the current evidence on the association between ISR and autoimmune diseases, focusing on pathogenic mechanisms and therapeutic targets. METHODS We conducted a comprehensive review of the literature on the relationship between ISR and insulin-dependent diabetes mellitus (IDDM), rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), antiphospholipid-antibodies syndrome (APS), inflammatory bowel diseases (IBD), and Hashimoto's thyroiditis (HT). RESULTS Patients affected with IDDM, RA, SLE, APS, IBD and HT proved to face higher rates of ISR compared to the general population. The endothelial dysfunction seems the principal common pathogenic pathway for ISR and is attributed to both the immune system disorder and the systemic inflammation. Some evidence suggested that methotrexate and anti-tumor necrosis factor treatments can be effective in reducing ISR, while antibodies against vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 showed to reduce neointimal hyperplasia in animal models. CONCLUSIONS Autoimmune diseases are a risk factor for ISR. The study of the potential cardiovascular benefits of the current therapies, mainly anti-inflammatory drugs, and the pursuit of innovative treatments appear of paramount interest.
Collapse
Affiliation(s)
- Martino Pepe
- Cardiovascular Diseases Section, Department of Emergency and Organ Transplantation (DETO), University of Bari, Piazza G. Cesare 11, Bari (BA), 70120, Italy.
| | - Gianluigi Napoli
- Cardiovascular Diseases Section, Department of Emergency and Organ Transplantation (DETO), University of Bari, Piazza G. Cesare 11, Bari (BA), 70120, Italy
| | - Eugenio Carulli
- Cardiovascular Diseases Section, Department of Emergency and Organ Transplantation (DETO), University of Bari, Piazza G. Cesare 11, Bari (BA), 70120, Italy
| | - Marco Moscarelli
- Cardiothoracic and Vascular Department, Maria Cecilia Hospital GVM Care & Research, Via Via Corriera 1,Cotignola, 48033, Ravenna, Italy
| | - Cinzia Forleo
- Cardiovascular Diseases Section, Department of Emergency and Organ Transplantation (DETO), University of Bari, Piazza G. Cesare 11, Bari (BA), 70120, Italy
| | - Palma Luisa Nestola
- Cardiovascular Diseases Section, Department of Emergency and Organ Transplantation (DETO), University of Bari, Piazza G. Cesare 11, Bari (BA), 70120, Italy
| | - Giuseppe Biondi-Zoccai
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Republica 79, Latina, 04100,Latina, Italy; Mediterranea Cardiocentro, Via Orazio 2, Napoli, 80122, Napoli, Italy
| | - Arturo Giordano
- Invasive Cardiology Unit, "Pineta Grande" Hospital, Via Domitiana km 30, Castel Volturno, 81030, Caserta, Italy
| | - Stefano Favale
- Cardiovascular Diseases Section, Department of Emergency and Organ Transplantation (DETO), University of Bari, Piazza G. Cesare 11, Bari (BA), 70120, Italy
| |
Collapse
|
28
|
Zhang S, Li P, Xin M, Jin X, Zhao L, Nan Y, Cheng XW. Dipeptidyl peptidase-4 inhibition prevents lung injury in mice under chronic stress via the modulation of oxidative stress and inflammation. Exp Anim 2021; 70:541-552. [PMID: 34219073 PMCID: PMC8614009 DOI: 10.1538/expanim.21-0067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Exposure to chronic psychosocial stress is a risk factor for various pulmonary diseases. In view of the essential role of dipeptidyl peptidase 4 (DPP4) in animal and human lung pathobiology, we investigated the role of DPP4 in stress-related lung injury in mice. Eight-week-old male mice were randomly divided into a non-stress group and a 2-week immobilization stress group. Non-stress control mice were left undisturbed. The mice subjected to immobilized stress were randomly assigned to the vehicle or the DPP4 inhibitor anagliptin for 2 weeks. Chronic stress reduced subcutaneous and inguinal adipose volumes and increased blood DPP4 levels. The stressed mice showed increased levels in the lungs of genes and/or proteins related to oxidative stress (p67phox, p47phox, p22phox and gp91phox), inflammation (monocyte chemoattractant protein-1, vascular cell adhesion molecule-1, and intracellular adhesion molecule-1), apoptosis (caspase-3, -8, -9), senescence (p16INK4A, p21, and p53) and proteolysis (matrix metalloproteinase-2 to -9, cathepsin S/K, and tissue inhibitor of matrix metalloproteinase-1 and -2), and reduced levels of eNOS, Sirt1, and Bcl-2 proteins; and these effects were reversed by genetic and pharmacological inhibitions of DPP4. We then exposed human umbilical vein endothelial cells in vitro to hydrogen peroxide; anagliptin treatment was also observed to mitigate oxidative and inflammatory molecules in this setting. Anagliptin can improve lung injury in stressed mice, possibly by mitigating vascular inflammation, oxidative stress production, and proteolysis. DPP4 may become a new therapeutic target for chronic psychological stress-related lung disease in humans and animals.
Collapse
Affiliation(s)
- Shengming Zhang
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| | - Ping Li
- State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union, Medical College
| | - Minglong Xin
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| | - Xianglan Jin
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| | - Longguo Zhao
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| | - Yongshan Nan
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| | - Xian Wu Cheng
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| |
Collapse
|
29
|
Deficiency of cysteinyl cathepsin K suppresses the development of experimental intimal hyperplasia in response to chronic stress. J Hypertens 2021; 38:1514-1524. [PMID: 32205563 DOI: 10.1097/hjh.0000000000002424] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Chronic psychological stress (CPS) is linked to cardiovascular disease initiation and progression. Given that cysteinyl cathepsin K (CatK) participates in vascular remodeling and atherosclerotic plaque growth in several animal models, we investigated the role of CatK in the development of experimental neointimal hyperplasia in response to chronic stress. METHODS AND RESULTS At first, male wild-type (CatK) mice that underwent carotid ligation injury were subjected to chronic immobilization stress. On postoperative and stressed day 14, the results demonstrated that stress accelerated injury-induced neointima hyperplasia. On day 4, stressed mice showed following: increased levels of monocyte chemoattractant protein-1, gp91phox, toll-like receptor-2 (TLR2), TLR4, and CatK mRNAs or/and proteins, oxidative stress production, aorta-derived smooth muscle cell (SMC) migration, and macrophage infiltration as well as targeted intracellular proliferating-related molecules. Stressed mice showed increased matrix metalloproteinase-2 (MMP-2) and MMP-9 mRNA expressions and activities and elastin disruption in the injured carotid arteries. Second, CatK and CatK deficiency (CatK) mice received ligation injury and stress to explore the role of CatK. The stress-induced harmful changes were prevented by CatK. Finally, CatK mice that had undergone ligation surgery were randomly assigned to one of two groups and administered vehicle or CatK inhibitor for 14 days. Pharmacological CatK intervention produced a vascular benefit. CONCLUSION These data indicate that CatK deletion protects against the development of experimental neointimal hyperplasia via the attenuation of inflammatory overaction, oxidative stress production, and VSMC proliferation, suggesting that CatK is a novel therapeutic target for the management of CPS-related restenosis after intravascular intervention therapies.
Collapse
|
30
|
Fisetin Alleviates Neointimal Hyperplasia via PPAR γ/PON2 Antioxidative Pathway in SHR Rat Artery Injury Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6625517. [PMID: 33968295 PMCID: PMC8084648 DOI: 10.1155/2021/6625517] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 03/18/2021] [Accepted: 04/09/2021] [Indexed: 11/17/2022]
Abstract
The phenotypic transformation of proliferation and migration in vascular smooth muscle cells (VSMCs) from media to intima is the basic pathology of neointimal hyperplasia after angioplasty in hypertensive patients. Angiotensin II (AngII) stimulates oxidative stress in VSMC, inducing VSMC proliferation and migration, which is a critical factor in both developments of hypertension and angioplasty-induced arterial restenosis. Fisetin, a plant flavonoid polyphenol, has been reported to be antioxidative and potent senolytic. It is unknown whether fisetin would inhibit neointimal hyperplasia. Therefore, we investigated the role of fisetin in neointimal formation in vitro and in vivo. The rat thoracic aortic smooth muscle cells (A10 cells) stimulated by AngII were used as the in vitro neointimal hyperplasia model, where AngII significantly induced the proliferation and migration in A10 cells. We found that fisetin could dose-dependently inhibit the effect of AngII via inducing the expression of an antioxidant, paraoxonase-2 (PON2), whose overexpression could inhibit the proliferation and migration of A10 cells and downexpression by siRNA had the opposite effect. Furthermore, we found the mechanism of fisetin's inducing PON2 expression involved PPARγ. Rosiglitazone, a PPARγ agonist, could increase PON2 expression in A10 cells, while the PPARγ inhibitor prevented the effect of fisetin on PON2. The in vivo neointimal hyperplasia model was established 2 weeks after the carotid artery balloon injury in SHR rats. Administration of fisetin (ip 3 mg/kg daily for 2 weeks) right after the injury significantly increased PON2 expression in the artery, inhibiting ROS production, and efficiently reduced carotid neointimal hyperplasia. These results indicate that fisetin increases the expression of antioxidant PON2 via activation of PPARγ, reducing oxidative stress, inhibiting VSMC proliferation and migration, and alleviates neointimal hyperplasia after intimal injury. PON2 may be a potential therapeutic target to reduce arterial remodeling after angioplasty in hypertensive patients.
Collapse
|
31
|
Zheng M, Guo J, Li Q, Yang J, Han Y, Yang H, Yu M, Zhong L, Lu D, Li L, Sun L. Syntheses and characterization of anti-thrombotic and anti-oxidative Gastrodin-modified polyurethane for vascular tissue engineering. Bioact Mater 2021; 6:404-419. [PMID: 32995669 PMCID: PMC7486448 DOI: 10.1016/j.bioactmat.2020.08.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 12/15/2022] Open
Abstract
Vascular grafts must avoid negative inflammatory responses and thrombogenesis to prohibit fibrotic deposition immediately upon implantation and promote the regeneration of small diameter blood vessels (<6 mm inner diameter). Here, polyurethane (PU) elastomers incorporating anti-coagulative and anti-inflammatory Gastrodin were fabricated. The films had inter-connected pores with porosities equal to or greater than 86% and pore sizes ranging from 250 to 400 μm. Incorporation of Gastrodin into PU films resulted in desirable mechanical properties, hydrophilicity, swelling ratios and degradation rates without collapse. The released Gastrodin maintained bioactivity over 21 days as assessed by its anti-oxidative capability. The Gastrodin/PU had better anti-coagulation response (less observable BSA, fibrinogen and platelet adhesion/activation and suppressed clotting in whole blood). Red blood cell compatibility, measured by hemolysis, was greatly improved with 2Gastrodin/PU compared to other Gastrodin/PU groups. Notably, Gastrodin/PU upregulated anti-oxidant factors Nrf2 and HO-1 expression in H2O2 treated HUVECs, correlated with decreasing pro-inflammatory cytokines TNF-α and IL-1β in RAW 264.7 cells. Upon implantation in a subcutaneous pocket, PU was encapsulated by an obvious fibrous capsule, concurrent with a large amount of inflammatory cell infiltration, while Gastrodin/PU induced a thinner fibrous capsule, especially 2Gastrodin/PU. Further, enhanced adhesion and proliferation of HUVECs seeded onto films in vitro demonstrated that 2Gastrodin/PU could help cell recruitment, as evidenced by rapid host cell infiltration and substantial blood vessel formation in vivo. These results indicate that 2Gastrodin/PU has the potential to facilitate blood vessel regeneration, thus providing new insight into the development of clinically effective vascular grafts.
Collapse
Affiliation(s)
- Meng Zheng
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Jiazhi Guo
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Qing Li
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Jian Yang
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yi Han
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Hongcai Yang
- Department of Neurology, The First Affiliated Hospital, Kunming Medical University, Kunming, 650500, China
| | - Mali Yu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Lianmei Zhong
- Department of Neurology, The First Affiliated Hospital, Kunming Medical University, Kunming, 650500, China
| | - Di Lu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Limei Li
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Lin Sun
- Department of Cardiology, The Second Affiliated Hospital, Kunming Medical University, Kunming, 650032, China
| |
Collapse
|
32
|
Watanabe Y, Mitomo S, Naganuma T, Chieffo A, Montorfano M, Nakamura S, Colombo A. Impact of Chronic Kidney Disease in Patients With Diabetes Mellitus after Percutaneous Coronary Intervention for Left Main Distal Bifurcation (From the Milan and New-Tokyo (MITO) Registry). Am J Cardiol 2021; 138:33-39. [PMID: 33058802 DOI: 10.1016/j.amjcard.2020.10.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 02/02/2023]
Abstract
The impact of chronic kidney disease (CKD) on clinical outcomes after percutaneous coronary intervention for unprotected left main distal bifurcation lesions in patients with diabetes mellitus (DM) is not fully understood in drug eluting stent era. We identified 512 consecutive DM patients who underwent percutaneous coronary intervention for unprotected left main distal bifurcation lesions at New Tokyo Hospital, San Raffaele Scientific Institute and EMO-GVM Centro Cuore Columbus between January 2005 and December 2015. We analyzed according to estimated glomerular filtration rate (eGFR). Each group was defined as follows; no CKD (60 ≤ eGFR), mild CKD (45 ≤ eGFR < 60), moderate CKD (30 ≤ eGFR < 45), and severe CKD (15 ≤ eGFR < 30). The primary end point was target lesion failure (TLF) at 3 years. TLF was defined as a composite of cardiac death, target lesion revascularization, and myocardial infarction. The rate of TLF was significantly higher in the severe CKD group than that in the other groups (Adjusted HR of severe CKD relative to the others 3.64, [1.86 to 7.11], p < 0.001). Cardiac mortality was significantly higher in the severe CKD group than that in the other groups (Adjusted HR of severe CKD relative to the others 6.43, [2.19 to 18.9], p = 0.001). Target lesion revascularization rate was comparable in 4 groups (Adjusted HR of severe CKD relative to the others 1.71, [0.60 to 4.82], p = 0.31). In conclusions, in DM patients, those with severe CKD was extremely associated with worse clinical outcomes.
Collapse
|
33
|
Gunawardena T, Merinopoulos I, Wickramarachchi U, Vassiliou V, Eccleshall S. Endothelial Dysfunction and Coronary Vasoreactivity - A Review of the History, Physiology, Diagnostic Techniques, and Clinical Relevance. Curr Cardiol Rev 2021; 17:85-100. [PMID: 32552654 PMCID: PMC8142375 DOI: 10.2174/1573403x16666200618161942] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/23/2020] [Accepted: 04/09/2020] [Indexed: 01/08/2023] Open
Abstract
The fervency for advancement and evolution in percutaneous coronary intervention has revolutionised the treatment of coronary artery disease. Historically, the focus of the interventional cardiologist was directed at the restoration of luminal patency of the major epicardial coronary arteries, yet whilst this approach is evolving with much greater utilisation of physiological assessment, it often neglects consideration of the role of the coronary microcirculation, which has been shown to clearly influence prognosis. In this review, we explore the narrative of the coronary circulation as more than just a simple conduit for blood but an organ with functional significance. We review organisation and physiology of the coronary circulation, as well as the current methods and techniques used to examine it. We discuss the studies exploring coronary artery endothelial function, appreciating that coronary artery disease occurs on a spectrum of disorder and that percutaneous coronary intervention has a latent effect on the coronary circulation with long-term consequences. It is concluded that greater recognition of the coronary artery endothelium and mechanisms of the coronary circulation should further guide revascularisation strategies.
Collapse
Affiliation(s)
- Tharusha Gunawardena
- Address correspondence to this author at the Department of Cardiology, Norfolk and Norwich University Hospital, Colney Lane NR4 7UY, Norwich, England; E-mail:
| | | | | | | | | |
Collapse
|
34
|
Interplay of pro-inflammatory cytokines, pro-inflammatory microparticles and oxidative stress and recurrent ventricular arrhythmias in elderly patients after coronary stent implantations. Cytokine 2020; 137:155345. [PMID: 33137563 DOI: 10.1016/j.cyto.2020.155345] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 09/09/2020] [Accepted: 10/12/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND The roles of pro-inflammatory microparticles, pro-inflammatory cytokines and oxidative stress were unknown in elderly patients with recurrent ventricular arrhythmias (VA). We evaluated whether cross talk between oxidative stress, pro-inflammatory microparticles, and pro-inflammatory cytokines play the roles in elderly patients with recurrent VA after coronary stenting. This research sought to investigate the effects of oxidative stress, pro-inflammatory microparticles, and pro-inflammatory cytokines on recurrent VA in elderly patients after coronary stenting. METHODS In this study, we included 613 consecutive elderly patients with recurrent ventricular arrhythmias induced by coronary reocclusions after coronary stenting. We measured CD31+ endothelial microparticle (CD31+EMP), CD62E+ endothelial microparticle (CD62E+EMP), high-sensitivity C-reactive protein (hs-CRP), aldosterone (ALD), malondialdehyde (MDA), tumor necrosis factor-α (TNF-α), soluble tumor necrosis factor receptor-1 (sTNFR-1) and soluble tumor necrosis factor receptor-2 (sTNFR-2) in elderly patients with recurrent VA and assessed impacts of pro-inflammatory microparticles, pro-inflammatory cytokines and oxidative stress on recurrent VA in elderly patients after coronary stenting. RESULTS The levels of CD31+EMP, CD62E+EMP, hs-CRP, ALD, MDA, TNF-α, sTNFR-1 and sTNFR-2 were increased in recurrent malignant ventricular arrhythmia, sustained ventricular tachycardia, multiple ventricular premature beat and left and right ventricular bundle branch block groups (P < 0.001) in elderly patients with coronary reocclusions after coronary stent implantation. Upregulation of pro-inflammatory microparticles, pro-inflammatory cytokines and oxidative stress markers induced recurrent VA in elderly patients after coronary stenting. CONCLUSIONS High levels of pro-inflammatory microparticles, pro-inflammatory cytokines and oxidative stress markers were associated with recurrent VA in elderly patients after coronary stenting. Our results suggested that the pro-inflammatory microparticles, pro-inflammatory cytokines and oxidative stress may simultaneously induce and aggravate recurrent VA in elderly patients after coronary stenting.
Collapse
|
35
|
Natural Drugs as a Treatment Strategy for Cardiovascular Disease through the Regulation of Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5430407. [PMID: 33062142 PMCID: PMC7537704 DOI: 10.1155/2020/5430407] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/02/2020] [Accepted: 09/09/2020] [Indexed: 02/07/2023]
Abstract
Oxidative stress (OS) refers to the physiological imbalance between oxidative and antioxidative processes leading to increased oxidation, which then results in the inflammatory infiltration of neutrophils, increased protease secretion, and the production of a large number of oxidative intermediates. Oxidative stress is considered an important factor in the pathogenesis of cardiovascular disease (CVD). At present, active components of Chinese herbal medicines (CHMs) have been widely used for the treatment of CVD, including coronary heart disease and hypertension. Since the discovery of artemisinin for the treatment of malaria by Nobel laureate Youyou Tu, the therapeutic effects of active components of CHM on various diseases have been widely investigated by the medical community. It has been found that various active CHM components can regulate oxidative stress and the circulatory system, including ginsenoside, astragaloside, and resveratrol. This paper reviews advances in the use of active CHM components that modulate oxidative stress, suggesting potential drugs for the treatment of various CVDs.
Collapse
|
36
|
Li CW, Li LL, Chen S, Zhang JX, Lu WL. Antioxidant Nanotherapies for the Treatment of Inflammatory Diseases. Front Bioeng Biotechnol 2020; 8:200. [PMID: 32258013 PMCID: PMC7093330 DOI: 10.3389/fbioe.2020.00200] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 02/28/2020] [Indexed: 12/11/2022] Open
Abstract
Reactive oxygen species (ROS) are essential in regulating various physiological functions. However, overproduction of ROS is implicated in the pathogenesis of various inflammatory diseases. Antioxidant therapy has thus represented an effective strategy for the treatment of oxidative stress relevant inflammatory diseases. Conventional anti-oxidative agents showed limited in vivo effects owing to their non-specific distribution and low retention in disease sites. Over the past decades, significant achievements have been made in the development of antioxidant nanotherapies that exhibit multiple advantages such as excellent pharmacokinetics, stable anti-oxidative activity, and intrinsic ROS-scavenging properties. This review provides a comprehensive overview on recent advances in antioxidant nanotherapies, including ROS-scavenging inorganic nanoparticles, organic nanoparticles with intrinsic antioxidant activity, and drug-loaded anti-oxidant nanoparticles. We highlight the biomedical applications of antioxidant nanotherapies in the treatment of different inflammatory diseases, with an emphasis on inflammatory bowel disease, cardiovascular disease, and brain diseases. Current challenges and future perspectives to promote clinical translation of antioxidant nanotherapies are also briefly discussed.
Collapse
Affiliation(s)
- Chen-Wen Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Lan-Lan Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China.,Department of Chemistry, College of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Sheng Chen
- Department of Pediatrics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jian-Xiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Wan-Liang Lu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
37
|
Moonikh KO, Kashef M, Mahmoudi K, Salehpour M. Effects of Quercetin Supplementation on Oxidative Stress, Blood Pressure, Aerobic Power, Concentric Pathologic Hypertrophy and Cardiac Function in Men with Hypertension and Coronary Artery Disease After Percutaneous Coronary Intervention: a Randomized, Double-Blind Placebo-Controlled Trial. NUTRITION AND FOOD SCIENCES RESEARCH 2020. [DOI: 10.29252/nfsr.7.2.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
38
|
Hu L, Huang Z, Ishii H, Wu H, Suzuki S, Inoue A, Kim W, Jiang H, Li X, Zhu E, Piao L, Zhao G, Lei Y, Okumura K, Shi GP, Murohara T, Kuzuya M, Cheng XW. PLF-1 (Proliferin-1) Modulates Smooth Muscle Cell Proliferation and Development of Experimental Intimal Hyperplasia. J Am Heart Assoc 2019; 8:e005886. [PMID: 31838975 PMCID: PMC6951060 DOI: 10.1161/jaha.117.005886] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Although apoptosis and cell proliferation have been extensively investigated in atherosclerosis and restenosis postinjury, the communication between these 2 cellular events has not been evaluated. Here, we report an inextricable communicative link between apoptosis and smooth muscle cell proliferation in the promotion of vascular remodeling postinjury. Methods and Results Cathepsin K-mediated caspase-8 maturation is a key initial step for oxidative stress-induced smooth muscle cell apoptosis. Apoptotic cells generate a potential growth-stimulating signal to facilitate cellular mass changes in response to injury. One downstream mediator that cathepsin K regulates is PLF-1 (proliferin-1), which can potently stimulate growth of surviving neighboring smooth muscle cells through activation of PI3K/Akt/p38MAPK (phosphatidylinositol 3-kinase/protein kinase B/p38 mitogen-activated protein kinase)-dependent and -independent mTOR (mammalian target of rapamycin) signaling cascades. We observed that cathepsin K deficiency substantially mitigated neointimal hyperplasia by reduction of Toll-like receptor-2/caspase-8-mediated PLF-1 expression. Interestingly, PLF-1 blocking, with its neutralizing antibody, suppressed neointima formation and remodeling in response to injury in wild-type mice. Contrarily, administration of recombinant mouse PLF-1 accelerated injury-induced vascular actions. Conclusions This is the first study detailing PLF-1 as a communicator between apoptosis and proliferation during injury-related vascular remodeling and neointimal hyperplasia. These data suggested that apoptosis-driven expression of PLF-1 is thus a novel target for treatment of apoptosis-based hyperproliferative disorders.
Collapse
Affiliation(s)
- Lina Hu
- Department of Public Health Guilin Medical College Guilin Guangxi China.,Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China.,Department of Community & Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan
| | - Zhe Huang
- Department of Neurology Occupational and Environmental Health Kitakyushu Hukuoka Japan
| | - Hideki Ishii
- Department of Cardiology Nagoya University Graduate School of Medicine Nagoya Japan
| | - Hongxian Wu
- Department of Cardiology Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Susumu Suzuki
- Department of Cardiology Nagoya University Graduate School of Medicine Nagoya Japan
| | - Aiko Inoue
- Department of Community & Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan.,Institute of Innovation for Future Society Nagoya University Nagoya Japan
| | - Weon Kim
- Division of Cardiology Department of Internal Medicine Kyung Hee University Seoul South Korea
| | - Haiying Jiang
- Department of Physiology and Pathophysiology Yanbian University School of Medicine Yanji Jinlin China
| | - Xiang Li
- Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China
| | - Enbo Zhu
- Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China
| | - Limei Piao
- Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China.,Department of Community & Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan
| | - Guangxian Zhao
- Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China
| | - Yanna Lei
- Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China
| | - Kenji Okumura
- Department of Cardiology Nagoya University Graduate School of Medicine Nagoya Japan
| | - Guo-Ping Shi
- Department of Cardiovascular Medicine Brigham and Women's Hospital and Harvard Medical School Boston MA
| | - Toyoaki Murohara
- Department of Cardiology Nagoya University Graduate School of Medicine Nagoya Japan
| | - Masafumi Kuzuya
- Department of Community & Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan.,Institute of Innovation for Future Society Nagoya University Nagoya Japan
| | - Xian Wu Cheng
- Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China.,Department of Community & Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan.,Division of Cardiology Department of Internal Medicine Kyung Hee University Seoul South Korea.,Institute of Innovation for Future Society Nagoya University Nagoya Japan
| |
Collapse
|
39
|
Xin M, Jin X, Cui X, Jin C, Piao L, Wan Y, Xu S, Zhang S, Yue X, Wang H, Nan Y, Cheng X. Dipeptidyl peptidase-4 inhibition prevents vascular aging in mice under chronic stress: Modulation of oxidative stress and inflammation. Chem Biol Interact 2019; 314:108842. [DOI: 10.1016/j.cbi.2019.108842] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 09/29/2019] [Indexed: 12/24/2022]
|
40
|
Xu X, Liu X, Yu L, Ma J, Yu S, Ni M. Impact of intracoronary nicorandil before stent deployment in patients with acute coronary syndrome undergoing percutaneous coronary intervention. Exp Ther Med 2019; 19:137-146. [PMID: 31853283 PMCID: PMC6909796 DOI: 10.3892/etm.2019.8219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 11/07/2019] [Indexed: 12/29/2022] Open
Abstract
The present study aimed to clarify the effect of bolus intracoronary nicorandil on inflammatory, oxidative and adherent indicators in patients with acute coronary syndrome (ACS) undergoing percutaneous coronary intervention (PCI). This randomized controlled trial (RCT) was performed to detect the inflammation and oxidative stress in intracoronary blood both before and after PCI. In total, 65 consecutive patients undergoing PCI were classified into a nicorandil therapy group (n=32) or a placebo group (n=33). All procedures were performed at Shandong University Qilu Hospital, China, during the period from March, 2016 to May, 2017. Intracoronary blood from patients who received nicorandil therapy during PCI showed no change in soluble CD40 ligand (sCD40L) concentration (1.86±0.08 vs. 1.90±0.09 ng/ml, P=0.12) but a significant increase was noted in the control group (1.87±0.17 vs. 2.82±0.26 ng/ml, P<0.01). This indicated a relative reduction in sCD40L level after PCI in the nicorandil group. We further demonstrated an increase in superoxide dismutase (SOD) activity (29.37±0.81 vs. 31.03±0.60 U/ml, P<0.001) and a reduction in lipid peroxidation (3.84±0.99 vs. 4.23±0.13 U/ml, P=0.001) in the nicorandil group but observed no change in the placebo group. ICAM-1 levels showed no change in the nicorandil group (69.54±6.89 vs. 72.01±8.25 ng/ml, P=0.83) but a significant increase in the control group after PCI in intracoronary blood (56.57±4.96 vs. 76.81±6.88 ng/ml, P=0.002). No changes were found in hs-CRP, TNFα and sVCAM-1 levels in coronary blood for both groups before and after PCI in ACS patients. Our findings demonstrate that intracoronary bolus nicorandil therapy has a significant effect on the inhibition of inflammatory indicators and oxidative stress in patients with ACS during PCI. This suggests a possible medical application of nicorandil for reducing inflammation and oxidative stress.
Collapse
Affiliation(s)
- Xingli Xu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xiaoling Liu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Liwen Yu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jing Ma
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Sufang Yu
- Department of Neurology, The Fourth People's Hospital, Liaocheng, Shandong 252002, P.R. China
| | - Mei Ni
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
41
|
Capturing Endothelial Cells by Coronary Stents - From Histology to Clinical Outcomes. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2019. [DOI: 10.2478/sjecr-2019-0018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Introduction of drug-eluting stents (DES) in the therapy of patients with coronary artery disease resulted in the significant reduction of in-stent restenosis compared to bare-metal stent (BMS) treatment. However, the high incidence of late stent thrombosis with DES emerged as one of the safety concerns after their implantation. Enhancing stent endothelization by improved early healing and neointimal strut coverage emerged as possible solution for this late complication. Endothelial progenitor cells (EPC) capturing stents are designed to promote in situ endothelization with immobilized, antihuman, anti-CD34 antibodies attached to the luminal stent surface. Anti-CD34 antibodies target and capture EPC from circulation, which further differentiate into vascular endothelial cells and form functional endothelial layer on the stent surface. These cells are also capable of secreting pro-angiogenic factors that stimulate local endothelial cells to proliferate and migrate. Preclinical and clinical studies proved feasibility, efficacy and safety of EPC capturing stents in stable and high-risk patients with coronary artery disease. Rapid and extensive endothelization of EPC capturing stents translated into favorable profile of clinical outcomes, comparable to efficacy of BMSs and DESs. Therefore, we here present the most important results from the experimental and clinical studies that explored ECP capturing strategy to enhance endothelization, reduce the incidence of instent thrombosis and improve outcomes of patients with coronary artery disease, along with the future perspectives in this promising therapeutic approach.
Collapse
|
42
|
Qu B, Yuan L, Yang L, Li J, Lv H, Yang X. Polyurethane End-Capped by Tetramethylpyrazine-Nitrone for Promoting Endothelialization Under Oxidative Stress. Adv Healthc Mater 2019; 8:e1900582. [PMID: 31529779 DOI: 10.1002/adhm.201900582] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 09/05/2019] [Indexed: 12/25/2022]
Abstract
Thrombus and restenosis are two main factors that cause the failure of vascular implants. Constructing a functional and confluent layer of endothelial cells (ECs) is considered an ideal method to prevent these problems. However, oxidative stress induced by the disease and implantation can damage ECs and hinder the endothelialization of implants. Thus, developing biomaterials that can protect ECs adhesion and proliferation from oxidative stress is urgently needed for the rapid endothelialization of vascular implants. In this work, a novel polyurethane (PU-TBN) is synthesized by employing tetramethylpyrazine-nitrone (TBN) as end-group to endow polymers with dual functions of antioxidant activity and promoting endothelialization. Common PU without TBN is also prepared to be control. Compared to PU, PU-TBN significantly promotes human umbilical vein endothelial cells (HUVECs) adhesion and proliferation, where cells spread well and a confluent endothelial layer is formed. PU-TBN also shows obvious free radical scavenging activity, and thus effectively attenuates oxidative stress to protect HUVECs from oxidative apoptosis. Moreover, PU-TBN exhibits enhanced antiplatelets effect, excellent biocompatibility, and similar mechanical properties to PU. These characteristics can endow PU-TBN with great potential to be used as vascular implants or coatings of other materials for rapid endothelialization under complex oxidative stress environment.
Collapse
Affiliation(s)
- Baoliu Qu
- State Key Laboratory of Polymer Physics and ChemistryChangchun Institute of Applied ChemistryChinese Academy of Sciences 5625 Renmin Stree Changchun 130022 P. R. China
- Polymer Composites Engineering LaboratoryChangchun Institute of Applied ChemistryUniversity of Chinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Liguang Yuan
- State Key Laboratory of Polymer Physics and ChemistryChangchun Institute of Applied ChemistryChinese Academy of Sciences 5625 Renmin Stree Changchun 130022 P. R. China
- Polymer Composites Engineering LaboratoryChangchun Institute of Applied ChemistryUniversity of Chinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Lei Yang
- State Key Laboratory of Polymer Physics and ChemistryChangchun Institute of Applied ChemistryChinese Academy of Sciences 5625 Renmin Stree Changchun 130022 P. R. China
- Polymer Composites Engineering LaboratoryChangchun Institute of Applied ChemistryUniversity of Chinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
- College of Applied Chemistry and EngineeringUniversity of Science and Technology of China 96 Jinzhai Road Hefei 230026 P. R. China
| | - Jinge Li
- State Key Laboratory of Polymer Physics and ChemistryChangchun Institute of Applied ChemistryChinese Academy of Sciences 5625 Renmin Stree Changchun 130022 P. R. China
- Polymer Composites Engineering LaboratoryChangchun Institute of Applied ChemistryUniversity of Chinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
| | - Hongying Lv
- State Key Laboratory of Polymer Physics and ChemistryChangchun Institute of Applied ChemistryChinese Academy of Sciences 5625 Renmin Stree Changchun 130022 P. R. China
- Polymer Composites Engineering LaboratoryChangchun Institute of Applied ChemistryUniversity of Chinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
| | - Xiaoniu Yang
- State Key Laboratory of Polymer Physics and ChemistryChangchun Institute of Applied ChemistryChinese Academy of Sciences 5625 Renmin Stree Changchun 130022 P. R. China
- Polymer Composites Engineering LaboratoryChangchun Institute of Applied ChemistryUniversity of Chinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
| |
Collapse
|
43
|
Polyhydroxyalkanoates based copolymers. Int J Biol Macromol 2019; 140:522-537. [PMID: 31437500 DOI: 10.1016/j.ijbiomac.2019.08.147] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/09/2019] [Accepted: 08/17/2019] [Indexed: 11/23/2022]
Abstract
Polyhydroxyalkanoates (PHAs) belong to a family of natural polyesters and are produced under unbalanced growth conditions as intracellular carbon and energy reserves by a wide variety of microorganisms. Being biodegradable, biocompatible and environmental friendly thermoplastics, the PHAs are considered as future polymers to replace petrochemicals based plastics. In this review, the introduction section deals with the brief discussion on PHA nature, availability, raw materials for production, processing etc. This is followed by the discussions on modifications. The copolymer syntheses by bacterial and chemical methods have been discussed. Under chemical methods, unsaturated side chains and their derivatives, oligomer, coupling, macro-initiating, trans-esterification, radiation grafting, click chemistry, ring opening and several miscellaneous polymerization methods have been elaborated. A brief discussion on applications has been incorporated. The last section includes conclusion and future perspectives.
Collapse
|
44
|
Wang H, Meng X, Piao L, Inoue A, Xu W, Yu C, Nakamura K, Hu L, Sasaki T, Wu H, Unno K, Umegaki H, Murohara T, Shi GP, Kuzuya M, Cheng XW. Cathepsin S Deficiency Mitigated Chronic Stress-Related Neointimal Hyperplasia in Mice. J Am Heart Assoc 2019; 8:e011994. [PMID: 31296090 PMCID: PMC6662117 DOI: 10.1161/jaha.119.011994] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background Exposure to chronic psychosocial stress is a risk factor for atherosclerosis-based cardiovascular disease. We previously demonstrated the increased expressions of cathepsin S (CatS) in atherosclerotic lesions. Whether CatS participates directly in stress-related neointimal hyperplasia has been unknown. Methods and Results Male wild-type and CatS-deficient mice that underwent carotid ligation injury were subjected to chronic immobilization stress for morphological and biochemical studies at specific times. On day 14 after stress/surgery, stress enhanced the neointima formation. At the early time points, the stressed mice had increased plaque elastin disruption, cell proliferation, macrophage accumulation, mRNA and/or protein levels of vascular cell adhesion molecule-1, angiotensin II type 1 receptor, monocyte chemoattractant protein-1, gp91phox, stromal cell-derived factor-1, C-X-C chemokine receptor-4, toll-like receptor-2, toll-like receptor-4, SC 35, galectin-3, and CatS as well as targeted intracellular proliferating-related molecules (mammalian target of rapamycin, phosphorylated protein kinase B, and p-glycogen synthase kinase-3α/β). Stress also increased the plaque matrix metalloproteinase-9 and matrix metalloproteinase-2 mRNA expressions and activities and aorta-derived smooth muscle cell migration and proliferation. The genetic or pharmacological inhibition of CatS by its specific inhibitor (Z- FL -COCHO) ameliorated the stressed arterial targeted molecular and morphological changes and stressed aorta-derived smooth muscle cell migration. Both the genetic and pharmacological interventions had no effect on increased blood pressure in stressed mice. Conclusions These results demonstrate an essential role of CatS in chronic stress-related neointimal hyperplasia in response to injury, possibly via the reduction of toll-like receptor-2/toll-like receptor-4-mediated inflammation, immune action, and smooth muscle cell proliferation, suggesting that CatS will be a novel therapeutic target for stress-related atherosclerosis-based cardiovascular disease.
Collapse
Affiliation(s)
- Hailong Wang
- 1 Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China.,2 Department of Community Health and Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan
| | - Xiangkun Meng
- 2 Department of Community Health and Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan
| | - Limei Piao
- 1 Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China.,2 Department of Community Health and Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan
| | - Aiko Inoue
- 2 Department of Community Health and Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan.,3 Institute of Innovation for Future Society Nagoya University Graduate School of Medicine Nagoya Japan
| | - Wenhu Xu
- 1 Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China.,2 Department of Community Health and Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan
| | - Chenglin Yu
- 1 Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China.,2 Department of Community Health and Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan
| | - Kae Nakamura
- 4 Department of Obstetrics and Gynecology Nagoya University Graduate School of Medicine Nagoya Japan
| | - Lina Hu
- 5 Department of Public Health Guilin Medical College Guangxi China
| | - Takeshi Sasaki
- 6 Department of Anatomy and Neuroscience Hamamatsu University School of Medicine Hamamatsu Japan
| | - Hongxian Wu
- 7 Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital Fudan University Shanghai China
| | - Kazumasa Unno
- 8 Department of Cardiology Nagoya University Graduate School of Medicine Nagoya Japan
| | - Hiroyuki Umegaki
- 2 Department of Community Health and Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan.,3 Institute of Innovation for Future Society Nagoya University Graduate School of Medicine Nagoya Japan
| | - Toyoaki Murohara
- 8 Department of Cardiology Nagoya University Graduate School of Medicine Nagoya Japan
| | - Guo-Ping Shi
- 9 Department of Medicine Brigham and Women's Hospital Harvard Medical School Boston MA
| | - Masafumi Kuzuya
- 2 Department of Community Health and Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan.,3 Institute of Innovation for Future Society Nagoya University Graduate School of Medicine Nagoya Japan
| | - Xian Wu Cheng
- 1 Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China.,2 Department of Community Health and Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan
| |
Collapse
|
45
|
Nanotherapies for Treatment of Cardiovascular Disease: A Case for Antioxidant Targeted Delivery. CURRENT PATHOBIOLOGY REPORTS 2019; 7:47-60. [PMID: 31396435 DOI: 10.1007/s40139-019-00196-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Purpose of Review Cardiovascular disease (CVD) involves a broad range of clinical manifestations resulting from a dysfunctional vascular system. Overproduction of reactive oxygen and nitrogen species are causally implicated in the severity of vascular dysfunction and CVD. Antioxidant therapy is an attractive avenue for treatment of CVD associated pathologies. Implementation of targeted nano-antioxidant therapies has the potential to overcome hurdles associated with systemic delivery of antioxidants. This review examines the currently available options for nanotherapeutic targeting CVD, and explores successful studies showcasing targeted nano-antioxidant therapy. Recent Findings Active targeting strategies in the context of CVD heavily focus on immunotargeting to inflammatory markers like cell adhesion molecules, or to exposed extracellular matrix components. Targeted antioxidant nanotherapies have found success in pre-clinical studies. Summary This review underscores the potential of targeted nanocarriers as means of finding success translating antioxidant therapies to the clinic, all with a focus on CVD.
Collapse
|
46
|
Tawa M, Shimosato T, Sakonjo H, Masuoka T, Nishio M, Ishibashi T, Okamura T. Chronological Change of Vascular Reactivity to cGMP Generators in the Balloon-Injured Rat Carotid Artery. J Vasc Res 2019; 56:109-116. [PMID: 31085923 DOI: 10.1159/000498896] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 02/13/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Soluble guanylate cyclase (sGC) exists as reduced, oxidized, and heme-free forms. Currently, it is unclear whether endovascular mechanical stenosis has an impact on vascular tone control by drugs targeting sGC, namely cGMP generators. METHODS Pharmacological responses to acidified sodium nitrite (reduced sGC stimulant) and BAY 60-2770 (oxidized/heme-free sGC stimulant) were studied in balloon-injured rat carotid arteries at several time points. In addition, sGC expression was detected by immunohistochemistry. RESULTS At 1 day after injury, acidified sodium nitrite-induced relaxation was attenuated in the injured artery, whereas BAY 60-2770-induced relaxation was augmented. Similar attenuation of response to acidified sodium nitrite was seen at 7 and 14 days after injury. On the other hand, the augmentation of response to BAY 60-2770 disappeared at 7 and 14 days after injury. At 1 day after injury, the immunohistochemical expression pattern of sGC in the smooth muscle layer of the injured artery was not different from that of the uninjured artery. However, in the injured artery, the intensity of sGC staining was weak at 7 and 14 days after injury. CONCLUSION Balloon injury alters vascular responsiveness to cGMP generators, which seems to be associated with the form and/or expression of sGC.
Collapse
Affiliation(s)
- Masashi Tawa
- Department of Pharmacology, Shiga University of Medical Science, Otsu, Japan, .,Department of Pharmacology, Kanazawa Medical University, Kahoku, Japan,
| | | | | | - Takayoshi Masuoka
- Department of Pharmacology, Kanazawa Medical University, Kahoku, Japan
| | - Matomo Nishio
- Department of Pharmacology, Kanazawa Medical University, Kahoku, Japan
| | | | - Tomio Okamura
- Department of Pharmacology, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
47
|
Jiang T, Xie Z, Wu F, Chen J, Liao Y, Liu L, Zhao A, Wu J, Yang P, Huang N. Hyaluronic Acid Nanoparticle Composite Films Confer Favorable Time-Dependent Biofunctions for Vascular Wound Healing. ACS Biomater Sci Eng 2019; 5:1833-1848. [PMID: 33405557 DOI: 10.1021/acsbiomaterials.9b00295] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Vascular stent implantation is the primary treatment for coronary artery disease. Surface modification of coronary stents is a topic of interest to prevent thrombosis and restenosis and to promote endothelization. However, bioactive coatings on implants have not yet been fully developed for the time-ordered biological requirements of vascular stents. The first month after vascular stent implantation, the pathological changes in the injured vascular tissue are complex and time-ordered. Therefore, vascular stents possess time-dependent biofunctions with early phase anticoagulant and anti-inflammatory properties. In the later stage, inhibitory effects on smooth muscle cell proliferation and the promotion of endothelial cell adhesion might meet the requirements of vascular repair. We fabricated three types of hyaluronic acid nanoparticles (HA-NPs) by subjecting HA and poly(ether imide) to ethyl(dimethylaminopropyl) carbodiimide/N-hydroxysuccinimide coupling reaction. The HA-NPs prepared by HA with a molecular weight of 100 kDa showed the best stability in a hyaluronidase environment. HA-NP composite films (HA-NCFs) were then fabricated by coimmobilizing selected HA-NPs (100 kDa) and HA molecules (100 kDa) through amide reaction on PDA/HD coated 316 L stainless steel surfaces. The detachment behavior of HA-NPs (100 kDa) in PBS for 20 days indicated that the HA-NPs (100 kDa) gradually detached from the surface. In vitro tests (anticoagulant and anti-inflammatory tests, endothelial cells, and smooth muscle cells seeding, and bacterial adhesion test) indicated that the newly fabricated HA-NCFs have inhibitory effects on the adhesion of fibrinogen, platelets, macrophages, bacteria, SMCs, and ECs. As the HA-NPs detached from the surface, the HA-NCFs showed excellent gradual comprehensive biocompatibility, which promoted adhesion and proliferation of ECs while still exerting inhibitory effects on the platelets, macrophages, and SMCs. Finally, in vivo SS wire implantation test (aortic implantation in healthy Sprague-Dawley rats) showed that HA-NCFs possessed anti-inflammatory properties, inhibited the proliferation of smooth muscle cells, and promoted re-endothelialization. In particular, HA-NCFs with time-dependent biofunctions showed better antirestenosis effects than those of surfaces modified with molecular HA, which exhibited constant biocompatibility. This study provides an important basis for the construction of HA-NP composite films with favorable time-dependent biofunctions for the time-ordered biological requirements of vascular stent.
Collapse
Affiliation(s)
- Ting Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, No. 111 of the North First Section of Second Ring Road, Chengdu 610031, PR China.,Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, No. 111 of the North First Section of Second Ring Road, Chengdu 610031, PR China
| | - Zhou Xie
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, No. 111 of the North First Section of Second Ring Road, Chengdu 610031, PR China
| | - Feng Wu
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, No. 111 of the North First Section of Second Ring Road, Chengdu 610031, PR China
| | - Jiang Chen
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, No. 111 of the North First Section of Second Ring Road, Chengdu 610031, PR China
| | - Yuzhen Liao
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, No. 111 of the North First Section of Second Ring Road, Chengdu 610031, PR China
| | - Luying Liu
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, No. 111 of the North First Section of Second Ring Road, Chengdu 610031, PR China
| | - Ansha Zhao
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, No. 111 of the North First Section of Second Ring Road, Chengdu 610031, PR China
| | - Jian Wu
- School of Life Science and Engineering, Southwest Jiaotong University, No. 111 of the North First Section of Second Ring Road, Chengdu 610031, PR China
| | - Ping Yang
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, No. 111 of the North First Section of Second Ring Road, Chengdu 610031, PR China
| | - Nan Huang
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, No. 111 of the North First Section of Second Ring Road, Chengdu 610031, PR China
| |
Collapse
|
48
|
The Role of Traditional Chinese Medicine in the Regulation of Oxidative Stress in Treating Coronary Heart Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3231424. [PMID: 30918578 PMCID: PMC6409025 DOI: 10.1155/2019/3231424] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 01/19/2019] [Accepted: 02/04/2019] [Indexed: 02/06/2023]
Abstract
Oxidative stress has been closely related with coronary artery disease. In coronary heart disease (CHD), an excess of reactive oxygen species (ROS) production generates endothelial cell and smooth muscle functional disorders, leading to a disequilibrium between the antioxidant capacity and prooxidants. ROS also leads to inflammatory signal activation and mitochondria-mediated apoptosis, which can promote and increase the occurrence and development of CHD. There are several kinds of antioxidative and small molecular systems of antioxidants, such as β-carotene, ascorbic acid, α-tocopherol, and reduced glutathione (GSH). Studies have shown that antioxidant treatment was effective and decreased the risk of CHD, but the effect of the treatment varies greatly. Traditional Chinese medicine (TCM) has been utilized for thousands of years in China and is becoming increasingly popular all over the world, especially for the treatments of cardiovascular diseases. This review will concentrate on the evidence of the action mechanism of TCM in preventing CHD by modulating oxidative stress-related signaling pathways.
Collapse
|
49
|
Tryfonos A, Green DJ, Dawson EA. Effects of Catheterization on Artery Function and Health: When Should Patients Start Exercising Following Their Coronary Intervention? Sports Med 2019; 49:397-416. [PMID: 30719682 DOI: 10.1007/s40279-019-01055-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Coronary artery disease (CAD) is a leading cause of death worldwide, and percutaneous transluminal coronary angiography (PTCA) and/or percutaneous coronary intervention (PCI; angioplasty) are commonly used to diagnose and/or treat the obstructed coronaries. Exercise-based rehabilitation is recommended for all CAD patients; however, most guidelines do not specify when exercise training should commence following PTCA and/or PCI. Catheterization can result in arterial dysfunction and acute injury, and given the fact that exercise, particularly at higher intensities, is associated with elevated inflammatory and oxidative stress, endothelial dysfunction and a pro-thrombotic milieu, performing exercise post-PTCA/PCI may transiently elevate the risk of cardiac events. This review aims to summarize extant literature relating to the impacts of coronary interventions on arterial function, including the time-course of recovery and the potential deleterious and/or beneficial impacts of acute versus long-term exercise. The current literature suggests that arterial dysfunction induced by catheterization recovers 4-12 weeks following catheterization. This review proposes that a period of relative arterial vulnerability may exist and exercise during this period may contribute to elevated event susceptibility. We therefore suggest that CAD patients start an exercise training programme between 2 and 4 weeks post-PCI, recognizing that the literature suggest there is a 'grey area' for functional recovery between 2 and 12 weeks post-catheterization. The timing of exercise onset should take into consideration the individual characteristics of patients (age, severity of disease, comorbidities) and the intensity, frequency and duration of the exercise prescription.
Collapse
Affiliation(s)
- Andrea Tryfonos
- Research Institute for Sport and Exercise Science, Liverpool John Moores University, Liverpool, L3 3AF, UK
| | - Daniel J Green
- School of Human Sciences (Exercise and Sport Science), The University of Western Australia, Crawley, WA, 6009, Australia
| | - Ellen A Dawson
- Research Institute for Sport and Exercise Science, Liverpool John Moores University, Liverpool, L3 3AF, UK.
| |
Collapse
|
50
|
Bhatia SK, Wadhwa P, Hong JW, Hong YG, Jeon JM, Lee ES, Yang YH. Lipase mediated functionalization of poly(3-hydroxybutyrate-co-3-hydroxyvalerate) with ascorbic acid into an antioxidant active biomaterial. Int J Biol Macromol 2019; 123:117-123. [DOI: 10.1016/j.ijbiomac.2018.11.052] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/06/2018] [Accepted: 11/10/2018] [Indexed: 01/01/2023]
|