1
|
Shi P, Sun P, Lou C, Fang J, Zhang L, Xie B, Zhang C. Adventitial Injection of HA/SA Hydrogel Loaded With IL-33 Antibody Decreases Neointimal Hyperplasia. J Surg Res 2024; 305:107-117. [PMID: 39667249 DOI: 10.1016/j.jss.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 09/30/2024] [Accepted: 11/18/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION Neointimal hyperplasia is one of the persistent complications after vascular interventions, and is the major cause of treatment failure. Interleukin-33 (IL-33) emerges as a crucial factor in many biological processes and plays an important role in vascular diseases. Adventitial injection is catching attention for its effectiveness and fewer side effects. We hypothesize that targeting IL-33 by adventitial injection can be a therapeutic method to inhibit neointimal hyperplasia. METHOD IL-33 expression was examined in human vein graft. The hydrogel was fabricated by the interaction of hyaluronic acid, sodium alginate, and CaCO3; and phosphate buffered saline (PBS) or IL-33 antibody or recombinant IL-33 was mixed within the hydrogel uniformly. A rat aortic wire injury-induced neointimal hyperplasia model was developed; rats were divided into three groups and received an adventitial injection of a hydrogel loaded with PBS or IL-33 antibody or recombinant IL-33 after wire injury. Tissues were harvested at day 21 and analyzed by histology and immunohistochemical staining. Hydrogel loaded with PBS, IL-33 antibody, or IL-33 was also used in a mouse carotid artery ligation neointimal hyperplasia model. RESULT There was a high expression of IL-33 in human vein graft neointima. Hydrogel can be successfully injected into the aortic wall and is encapsulated by the adventitia. The hydrogel could be seen beneath the adventitia after adventitial injection and was partly degraded at day 21. There was a significantly thinner neointimal thickness and less proliferation and inflammation in the IL-33 antibody group compared to the control group. On the contrary, the IL-33 group has a thicker neointima, increased proliferation, and inflammation. The mouse carotid artery ligation model showed similar results. CONCLUSIONS IL-33 plays a role in arterial neointimal hyperplasia in both human and rodent models; adventitial injection of hydrogel loaded with IL-33 antibody can effectively decrease neointimal thickness. Neutralizing IL-33 by IL-33 antibody may be a potential therapeutic method to inhibit intimal hyperplasia after vascular interventions.
Collapse
Affiliation(s)
- Pengfei Shi
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Peng Sun
- Key Vascular Physiology and Applied Research Laboratory of Zhengzhou City, Zhengzhou, China; Department of Cardiovascular Surgery Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vascular Diseases, Beijing, China
| | - Chunyang Lou
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianbang Fang
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liwei Zhang
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Boao Xie
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Cong Zhang
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
2
|
Liu Y, Huang T, Yap NA, Lim K, Ju LA. Harnessing the power of bioprinting for the development of next-generation models of thrombosis. Bioact Mater 2024; 42:328-344. [PMID: 39295733 PMCID: PMC11408160 DOI: 10.1016/j.bioactmat.2024.08.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024] Open
Abstract
Thrombosis, a leading cause of cardiovascular morbidity and mortality, involves the formation of blood clots within blood vessels. Current animal models and in vitro systems have limitations in recapitulating the complex human vasculature and hemodynamic conditions, limiting the research in understanding the mechanisms of thrombosis. Bioprinting has emerged as a promising approach to construct biomimetic vascular models that closely mimic the structural and mechanical properties of native blood vessels. This review discusses the key considerations for designing bioprinted vascular conduits for thrombosis studies, including the incorporation of key structural, biochemical and mechanical features, the selection of appropriate biomaterials and cell sources, and the challenges and future directions in the field. The advancements in bioprinting techniques, such as multi-material bioprinting and microfluidic integration, have enabled the development of physiologically relevant models of thrombosis. The future of bioprinted models of thrombosis lies in the integration of patient-specific data, real-time monitoring technologies, and advanced microfluidic platforms, paving the way for personalized medicine and targeted interventions. As the field of bioprinting continues to evolve, these advanced vascular models are expected to play an increasingly important role in unraveling the complexities of thrombosis and improving patient outcomes. The continued advancements in bioprinting technologies and the collaboration between researchers from various disciplines hold great promise for revolutionizing the field of thrombosis research.
Collapse
Affiliation(s)
- Yanyan Liu
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Tao Huang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Nicole Alexis Yap
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Khoon Lim
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- School of Medical Sciences, The University of Sydney, Darlington, NSW 2008, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW 2042, Australia
| |
Collapse
|
3
|
Guo Z, Zhang Y, Peng Z, Rao H, Yang J, Chen Z, Song W, Wan Q, Chen H, Wang M. Complement factor B, not the membrane attack complex component C9, promotes neointima formation after arterial wire injury. Atherosclerosis 2024; 399:118586. [PMID: 39500113 DOI: 10.1016/j.atherosclerosis.2024.118586] [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: 02/09/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND AND AIMS Vascular restenosis due to neointima hyperplasia limits the long-term patency of stented arteries, resulting in angioplasty failure. The complement system has been implicated in restenosis. This study aimed to investigate the role of complement factor B (fB), an essential component of the alternative pathway of complement activation, in neointima formation. METHODS Angioplasty wire injury was conducted using 12-week-old mice deficient in fB or C9 (the main component of the membrane attacking complex, C5b-9) and littermate controls and neointima formation were assessed. Vascular smooth muscle cell (SMC) and endothelial cell (EC) proliferation and migration were examined in vitro. RESULTS fB was mainly detected in SMCs of stenotic arteries from humans and mice. Deletion of fB substantially reduced the neointima area and intima-to-media area ratio without affecting the media area at 28 days after injury. At 7 days after injury, fB deficiency decreased SMC proliferation, unaltering neointimal macrophage infiltration and EC reendothelialization. Vascular SMC-expressed fB, not the circulation-sourced fB, played an essential role in SMC proliferation and migration in vitro. fB deficient mice exhibited lower levels of the soluble form of C5b-9, however, deletion of C9 did not alter neointima formation after wire injury, consistent with the null impact of C9 deficiency on SMC proliferation in vitro. CONCLUSIONS fB promotes neointima formation following wire-induced artery injury independent of forming the membrane-attacking complex. This is attributable to fB-dependent SMC proliferation and migration without affecting EC function. Targeting fB might protect against restenosis after percutaneous coronary intervention.
Collapse
MESH Headings
- Animals
- Neointima
- Cell Proliferation
- Cell Movement
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Humans
- Mice, Knockout
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/injuries
- Mice, Inbred C57BL
- Disease Models, Animal
- Mice
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Male
- Complement Membrane Attack Complex/metabolism
- Cells, Cultured
- Vascular System Injuries/pathology
- Vascular System Injuries/genetics
- Vascular System Injuries/metabolism
Collapse
Affiliation(s)
- Ziyi Guo
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Yuze Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Zekun Peng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Haojie Rao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Jianfeng Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Zengrong Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Wenchao Song
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Qing Wan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Hong Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Miao Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China; Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing, 100037, China; National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China.
| |
Collapse
|
4
|
Chang GJ, Chen WJ, Hsu YJ, Chen YH. Empagliflozin Attenuates Neointima Formation After Arterial Injury and Inhibits Smooth Muscle Cell Proliferation and Migration by Suppressing Platelet-Derived Growth Factor-Related Signaling. J Am Heart Assoc 2024; 13:e035044. [PMID: 39508166 DOI: 10.1161/jaha.124.035044] [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: 02/17/2024] [Accepted: 07/23/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Sodium-glucose cotransporter 2 (SGLT2) inhibitors reduce cardiovascular events. However, the precise mechanisms beyond glycemic control are not fully understood. The objective of this study was to determine the role of PDGF (platelet-derived growth factor)-related signaling in empagliflozin-mediated inhibition of neointima formation. METHODS AND RESULTS Adult male nondiabetic Wistar rats were subjected to carotid artery balloon injury. Empagliflozin (30 mg/kg per day) was administered by oral gavage for 18 days beginning 4 days before surgery. The in vitro effects of empagliflozin on rat aortic vascular smooth muscle cell (VSMC) proliferation and migration were also determined. Empagliflozin attenuated balloon injury-induced neointima formation in carotid arteries. In VSMCs, empagliflozin attenuated PDGF-BB-induced proliferation and migration. Moreover, empagliflozin-treated VSMCs did not undergo apoptosis or cytotoxic death. Empagliflozin suppressed PDGF-related signaling, including phosphorylation of PDGF receptor β, Akt, and STAT3 (signal transducer and activator of transcription 3). Overactivation of PDGF signaling attenuated empagliflozin-mediated inhibition of VSMC function. SGLT2 mRNA levels in rat VSMCs were undetectable, and SGLT2 silencing did not alter the empagliflozin-mediated effects, supporting the SGLT2-independent effects of empagliflozin on VSMC. CONCLUSIONS This study highlights the crucial role of suppressing PDGF-related signaling in mediating the beneficial effects of empagliflozin on neointima formation and VSMC function, which are independent of SGLT2 and glycemic control. Our study provides a novel mechanistic aspect of empagliflozin for the prevention of vascular stenosis disorders.
Collapse
MESH Headings
- Animals
- Glucosides/pharmacology
- Male
- Neointima
- Benzhydryl Compounds/pharmacology
- Cell Proliferation/drug effects
- Cell Movement/drug effects
- Signal Transduction/drug effects
- Carotid Artery Injuries/pathology
- Carotid Artery Injuries/drug therapy
- Carotid Artery Injuries/metabolism
- Rats, Wistar
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Sodium-Glucose Transporter 2 Inhibitors/pharmacology
- Cells, Cultured
- Disease Models, Animal
- Phosphorylation
- STAT3 Transcription Factor/metabolism
- Rats
- Proto-Oncogene Proteins c-akt/metabolism
- Becaplermin/pharmacology
- Becaplermin/metabolism
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Platelet-Derived Growth Factor/metabolism
- Platelet-Derived Growth Factor/pharmacology
Collapse
Affiliation(s)
- Gwo-Jyh Chang
- Graduate Institute of Clinical Medicinal Sciences Chang-Gung University College of Medicine Tao-Yuan Taiwan
- Cardiovascular Division, Chang-Gung Memorial Hospital Chang-Gung University College of Medicine Tao-yuan Taiwan
| | - Wei-Jan Chen
- Cardiovascular Division, Chang-Gung Memorial Hospital Chang-Gung University College of Medicine Tao-yuan Taiwan
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital National Defense Medical Center Taipei Taiwan
| | - Ying-Hwa Chen
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital National Yang Ming Chiao Tung University College of Medicine Taipei Taiwan
| |
Collapse
|
5
|
Wu H, Li D, Zhang CY, Huang LL, Zeng YJ, Chen TG, Yu K, Meng JW, Lin YX, Guo R, Zhou Y, Gao G. Restoration of ARA metabolic disorders in vascular smooth muscle cells alleviates intimal hyperplasia. Eur J Pharmacol 2024; 983:176824. [PMID: 39265882 DOI: 10.1016/j.ejphar.2024.176824] [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: 04/06/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 09/14/2024]
Abstract
Intimal hyperplasia (IH) is an innegligible issue for patients undergoing interventional therapy. The proliferation and migration of vascular smooth muscle cells (VSMCs) induced by platelet-derived growth factor-BB (PDGF-BB) are critical events in the development of IH. While the exact mechanism and effective target for IH needs further investigation. Metabolic disorders of arachidonic acid (ARA) are involved in the occurrence and progression of various diseases. In this study, we found that the expressions of soluble epoxide hydrolase (sEH) and cyclooxygenase-2 (COX-2) were significantly increased in the VSMCs during balloon injury-induced IH. Then, we employed a COX-2/sEH dual inhibitor PTUPB to increase the concentration of epoxyeicosatrienoic acids (EETs) while prevent the release of pro-inflammatory prostaglandins. Results showed that PTUPB treatment significantly reduced neointimal thickening induced by balloon injury in rats in vivo and inhibited PDGF-BB-induced proliferation and migration of VSMCs in vitro. Our results showed that PTUPB may reverse the phenotypic transition of VSMCs by inhibiting Pttg1 expression. In conclusion, we found that the dysfunction of ARA metabolism in VSMCs contributes to IH, and the COX-2/sEH dual inhibitor PTUPB attenuates IH progression by reversing the phenotypic switch in VSMC through the Sirt1/Pttg1 pathway.
Collapse
MESH Headings
- Animals
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Hyperplasia
- Male
- Rats
- Cyclooxygenase 2/metabolism
- Cell Proliferation/drug effects
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Rats, Sprague-Dawley
- Cell Movement/drug effects
- Epoxide Hydrolases/antagonists & inhibitors
- Epoxide Hydrolases/metabolism
- Tunica Intima/pathology
- Tunica Intima/metabolism
- Tunica Intima/drug effects
- Becaplermin/pharmacology
- Neointima/pathology
- Neointima/metabolism
- Neointima/drug therapy
- Metabolic Diseases/metabolism
- Metabolic Diseases/drug therapy
- Metabolic Diseases/pathology
Collapse
Affiliation(s)
- Hui Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Dai Li
- Phase I Clinical Research Center, Xiangya Hospital, Central South University, Changsha, 410005, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410005, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410078, China
| | - Ling-Li Huang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - You-Jie Zeng
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Tian-Ge Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410005, China
| | - Ke Yu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Jia-Wei Meng
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Yu-Xin Lin
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Ren Guo
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410078, China.
| | - Ge Gao
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| |
Collapse
|
6
|
Zhao Y, Cui H, Guo X, Lang J. Biomechanical analysis of nickel-titanium (NiTi)-cobalt-chromium (CoCr) hybrid-braided dense-mesh stents for carotid artery stenosis. Comput Methods Biomech Biomed Engin 2024:1-12. [PMID: 39543441 DOI: 10.1080/10255842.2024.2428720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/12/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
This study investigates NiTi-CoCr hybrid carotid artery stents to enhance mechanical properties over NiTi-only designs. Different configurations (24NiTi, 20NiTi-4CoCr, 16NiTi-8CoCr, and 12NiTi-12CoCr) were evaluated through radial compression and bending simulations. The 12NiTi-12CoCr stent showed the highest radial support (39.37 N) and increased bending strength by 77.96%. When modeled in a stenotic artery, this stent reduced stenosis from 81.52% to 29.33% and improved blood flow dynamics, alleviating high-pressure zones and balancing wall shear stress. These results suggest that CoCr wires improve stent performance, with the 12NiTi-12CoCr stent offering significant biomechanical and hemodynamic benefits.
Collapse
Affiliation(s)
- Yunchuan Zhao
- Shanghai Institute for Minimally Invasive Therapy, University of Shanghai for Science and Technology, Shanghai, PR China
| | - Haipo Cui
- Shanghai Institute for Minimally Invasive Therapy, University of Shanghai for Science and Technology, Shanghai, PR China
| | - Xudong Guo
- Shanghai Institute for Minimally Invasive Therapy, University of Shanghai for Science and Technology, Shanghai, PR China
| | - Jingcheng Lang
- Shanghai Institute for Minimally Invasive Therapy, University of Shanghai for Science and Technology, Shanghai, PR China
| |
Collapse
|
7
|
Luo Y, Gu G, Li Y, Zheng B, Ren F, Wang J, Chen C, Chen Z, Zhang Y, Zhao B, Yang J, Wang J. A novel method to detect carotid artery in-stent restenosis. J Neuroimaging 2024; 34:664-672. [PMID: 39462837 DOI: 10.1111/jon.13245] [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: 08/21/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND AND PURPOSE Carotid artery stenosis is a major risk factor for ischemic stroke. Despite carotid artery stenting, in-stent restenosis (ISR) remains challenging. Pigs serve as an ideal ISR model. This study aims to establish a novel porcine model of carotid ISR using open-loop and closed-loop stents and to assess ISR with optical coherence tomography (OCT) and histopathology, comparing incidence and vascular response between stent types. METHODS Twelve adult male Bama miniature pigs underwent carotid stenting with either open-loop or closed-loop stents. The animals received antiplatelet therapy pre- and postimplantation. Postimplantation evaluations at 90 days included carotid digital subtraction angiography (DSA), OCT, histopathological examination, and electron microscopy. RESULTS Both stent types showed ISR as detected by OCT and DSA. OCT revealed comparable neointimal proliferation within stent struts for both types, with no significant differences in stent, lumen, and neointimal dimensions. Histopathological analysis and electron microscopy provided insights into tissue responses and healing processes following stent implantation. No significant difference in ISR incidence was found between the stent types based on a χ2 test (p = .110). OCT and hematoxylin-eosin staining exhibit the highest consistency in evaluating neointimal area. CONCLUSIONS The novel porcine ISR model demonstrated similar ISR outcomes for open-loop and closed-loop stents. OCT proved to be a highly consistent and valuable tool for evaluating stent and arterial conditions, comparable to histopathological findings. However, due to a small sample size, the validity of these preliminary findings requires further investigation to be confirmed.
Collapse
Affiliation(s)
- Yuding Luo
- Department of Neurology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Department of Neurology, Ya'an People's Hospital, Ya'an, China
| | - Gangfeng Gu
- Department of Neurology, Ya'an People's Hospital, Ya'an, China
| | - Yan Li
- Department of Neurology, Ya'an People's Hospital, Ya'an, China
- North Sichuan Medical College, Nanchong, China
| | - Bo Zheng
- Department of Neurology, Ya'an People's Hospital, Ya'an, China
| | - Fanzhou Ren
- Department of Neurology, Ya'an People's Hospital, Ya'an, China
- North Sichuan Medical College, Nanchong, China
| | - Junqiu Wang
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Chuanli Chen
- Department of Neurology, Ya'an People's Hospital, Ya'an, China
- North Sichuan Medical College, Nanchong, China
| | - Zhao Chen
- Department of Neurology, Ya'an People's Hospital, Ya'an, China
| | - Yingqian Zhang
- West China Hospital of Sichuan University, Chengdu, China
| | - Bangcheng Zhao
- West China Hospital of Sichuan University, Chengdu, China
| | - Jian Yang
- Science and Education Department, Ya'an People's Hospital, Ya'an, China
| | - Jian Wang
- Department of Neurology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Department of Neurology, Ya'an People's Hospital, Ya'an, China
| |
Collapse
|
8
|
Matsushita K, Sato C, Bruckert C, Gong D, Amissi S, Hmadeh S, Fakih W, Remila L, Lessinger JM, Auger C, Jesel L, Ohlmann P, Kauffenstein G, Schini-Kerth VB, Morel O. Potential of dapagliflozin to prevent vascular remodeling in the rat carotid artery following balloon injury. Atherosclerosis 2024; 397:117595. [PMID: 38879387 DOI: 10.1016/j.atherosclerosis.2024.117595] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND AND AIMS Sodium-glucose co-transporter 2 (SGLT2) inhibitors have been shown to reduce the risk of cardiovascular events independently of glycemic control. However, the possibility that SGLT2 inhibitors improve vascular restenosis is unknown. The aim of this study was to examine whether dapagliflozin could prevent neointima thickening following balloon injury and, if so, to determine the underlying mechanisms. METHODS Saline, dapagliflozin (1.5 mg/kg/day), or losartan (30 mg/kg/day) was administered orally for five weeks to male Wistar rats. Balloon injury of the left carotid artery was performed a week after starting the treatment and rats were sacrificed 4 weeks later. The extent of neointima was assessed by histomorphometric and immunofluorescence staining analyses. Vascular reactivity was assessed on injured and non-injured carotid artery rings, changes of target factors by immunofluorescence, RT-qPCR, and histochemistry. RESULTS Dapagliflozin and losartan treatments reduced neointima thickening by 32 % and 27 %, respectively. Blunted contractile responses to phenylephrine and relaxations to acetylcholine and down-regulation of eNOS were observed in the injured arteries. RT-qPCR investigations indicated an increased in gene expression of inflammatory (IL-1beta, VCAM-1), oxidative (p47phox, p22phox) and fibrotic (TGF-beta1) markers in the injured carotid. While these changes were not affected by dapagliflozin, increased levels of AT1R and NTPDase1 (CD39) and decreased levels of ENPP1 were observed in the restenotic carotid artery of the dapagliflozin group. CONCLUSIONS Dapagliflozin effectively reduced neointimal thickening. The present data suggest that dapagliflozin prevents restenosis through interfering with angiotensin and/or extracellular nucleotides signaling. SGLT2 represents potential new target for limiting vascular restenosis.
Collapse
Affiliation(s)
- Kensuke Matsushita
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France; Université de Strasbourg, Pôle D'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Strasbourg, France
| | - Chisato Sato
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France; Université de Strasbourg, Pôle D'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Strasbourg, France
| | - Christophe Bruckert
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - DalSeong Gong
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - Said Amissi
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - Sandy Hmadeh
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - Walaa Fakih
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - Lamia Remila
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - Jean-Marc Lessinger
- CHU de Strasbourg, Laboratoire de Biochimie Clinique et Biologie Moléculaire, 67091, Strasbourg, France
| | - Cyril Auger
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - Laurence Jesel
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France; Université de Strasbourg, Pôle D'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Strasbourg, France
| | - Patrick Ohlmann
- Université de Strasbourg, Pôle D'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Strasbourg, France
| | - Gilles Kauffenstein
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - Valérie B Schini-Kerth
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - Olivier Morel
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France; Université de Strasbourg, Pôle D'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Strasbourg, France; Hanoï Medical University, Hanoi, Viet Nam.
| |
Collapse
|
9
|
Xie L, Mao T, Gao Q, Pan Y, Yang Z, Qu X, Feng R, Xia J, Lin Q, Wan J. Comparative efficacy of the five most common traditional Chinese medicine monomers in reducing intimal hyperproliferation in arterial balloon injury models: A network meta-analysis. Heliyon 2024; 10:e36327. [PMID: 39263082 PMCID: PMC11387273 DOI: 10.1016/j.heliyon.2024.e36327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 09/13/2024] Open
Abstract
Objective This study utilized network meta-analysis (NMA) to compare the efficacy of five commonly used traditional Chinese medicine monomers in reducing intimal hyperproliferation in arterial balloon injury models. Methods Relevant literature up to January 2024 was systematically retrieved from seven major databases. The intima-to-media (I/M) ratio was chosen as the primary outcome measure. The risk of bias in animal studies was assessed using the SYstematic Review Centre for Laboratory Animal Experimentation (SYRCLE) tool. Statistical analysis was conducted using Stata 17 software. Results A total of 43 studies were included in this meta-analysis. NMA results showed that in the rat model, compared to the control group, GS (SMD: 0.99, 95%CI: 1.25 to -0.73), ASIV (SMD: 1.16, 95%CI: 1.65 to -0.67), TMP (SMD: 0.68, 95%CI: 1.31 to -0.05), and TPNS (SMD: 1.36, 95%CI: 1.91 to -0.80) exhibited inhibitory effects on postoperative intimal hyperproliferation, reducing the I/M ratio. In the rabbit model, compared to the control group, TPNS (SMD: 1.23, 95%CI: 1.97 to -0.49) inhibited postoperative intimal hyperproliferation and reduced the I/M ratio. Superiority ranking analysis suggested that total Panax notoginseng saponin (TPNS) might be the most effective traditional Chinese medicine monomer in reducing intimal hyperproliferation in arterial balloon injury models, lowering the I/M ratio. Conclusion NMA indicates that traditional Chinese medicine monomers can effectively reduce postoperative intimal hyperproliferation in arterial balloon injury models, lowering the I/M ratio, with TPNS showing optimal efficacy. However, the research on TIIA is insufficient, and the limited sample size may affect the robustness of the results. Furthermore, the majority of research on traditional Chinese medicine monomers is currently limited to the experimental stage, lacking further clinical validation. Conducting standardized animal experiments and reporting their findings can enhance the quality of evidence from animal studies, laying the foundation for future clinical trials.
Collapse
Affiliation(s)
- Long Xie
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Tianshi Mao
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qun Gao
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yi Pan
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhifei Yang
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xinyan Qu
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ruli Feng
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Junyan Xia
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qian Lin
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jie Wan
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, 100029, China
| |
Collapse
|
10
|
Weiss R, Yu Q, Funaki B, Hammes M. Recurrent thrombosis and patency of the arteriovenous access in patients receiving hemodialysis. J Vasc Access 2024:11297298241260755. [PMID: 39126205 DOI: 10.1177/11297298241260755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Thrombosis of the vascular access in patients with end-stage renal disease requiring hemodialysis are common and require timely interventional procedures to restore patency. The aim of the current study was to identify factors having a significant effect on patency rates after access thrombosis. Our hypothesis was the length of time between the initial clotting of the access and the subsequent percutaneous declotting impacts the patency rates of the vascular access. METHODS In this retrospective cohort study, patients with a clotted arteriovenous access between Jan 1, 2011, and Jan 1, 2016, were included. Demographics, access history, and associated details of the access procedure were reviewed from the electronic medical record. Statistical analysis was done using t-test and chi-square or fisher exact tests to compare arteriovenous fistulae (AVF) and arteriovenous grafts (AVG). Primary patency, defined as the time from index procedure to endpoint, was analyzed using the Kaplan-Meier method and log rank test. RESULTS There were 883 percutaneous declotting procedures reviewed. About 351 procedures were performed in patients with an AVF and 532 with an AVG. The mean time from thrombosis to declotting was 1.71 ± 2.29 days. The overall median primary patency for both AVF and AVG was 43 days with no difference in patency between patients with AVF (39 days) versus AVG (42 days; p = 0.385). The time period from access thrombosis to declotting did not affect patency rates for either AVG or AVF (p = 0.385). On multivariable analysis, prior intervention (HR: 1.32, 95% CI: 1.14-1.53, p < 0.001) and cardiovascular disease (HR: 1.19, 95% CI: 1.03-1.37, p = 0.016) were independently associated with access patency. CONCLUSIONS Time from thrombosis to declotting did not affect patency rates however once there was a thrombotic event, recurrent thrombosis requiring intervention was common with patency significantly decreased. Future prospective studies to validate our results and study pathogenic mechanisms of recurrent thrombosis are warranted.
Collapse
Affiliation(s)
- Ryan Weiss
- Department of Internal Medicine, Section of Nephrology, University of Chicago Medical Center, Chicago, IL, USA
| | - Qian Yu
- Department of Radiology, Section of Vascular and Interventional Radiology, University of Chicago Medical Center, Chicago, IL, USA
| | - Brian Funaki
- Department of Radiology, Section of Vascular and Interventional Radiology, University of Chicago Medical Center, Chicago, IL, USA
| | - Mary Hammes
- Department of Internal Medicine, Section of Nephrology, University of Chicago Medical Center, Chicago, IL, USA
| |
Collapse
|
11
|
Wen L, Fu X, Zhang H, Ye P, Fu H, Zhou Z, Sun R, Xu T, Fu C, Zhu C, Guo Y, Fan H. Tailoring Zinc Ferrite Nanoparticle Surface Coating for Macrophage-Affinity Magnetic Resonance Imaging of Atherosclerosis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:13496-13508. [PMID: 38449094 DOI: 10.1021/acsami.3c17212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the formation of atherosclerotic plaques, while macrophages as key players in plaque progression and destabilization are promising targets for atherosclerotic plaque imaging. Contrast-enhanced magnetic resonance imaging (CE-MRI) has emerged as a powerful noninvasive imaging technique for the evaluation of atherosclerotic plaques within arterial walls. However, the visualization of macrophages within atherosclerotic plaques presents considerable challenges due to the intricate pathophysiology of the disease and the dynamic behavior of these cells. Biocompatible ferrite nanoparticles with diverse surface ligands possess the potential to exhibit distinct relaxivity and cellular affinity, enabling improved imaging capabilities for macrophages in atherosclerosis. In this work, we report macrophage-affinity nanoparticles for magnetic resonance imaging (MRI) of atherosclerosis via tailoring nanoparticle surface coating. The ultrasmall zinc ferrite nanoparticles (Zn0.4Fe2.6O4) as T1 contrast agents were synthesized and modified with dopamine, 3,4-dihydroxyhydrocinnamic acid, and phosphorylated polyethylene glycol to adjust their surface charges to be positively, negatively, and neutrally charged, respectively. In vitro MRI evaluation shows that the T1 relaxivity for different surface charged Zn0.4Fe2.6O4 nanoparticles was three higher than that of the clinically used Gd-DTPA. Furthermore, in vivo atherosclerotic plaque MR imaging indicates that positively charged Zn0.4Fe2.6O4 showed superior MRI efficacy on carotid atherosclerosis than the other two, which is ascribed to high affinity to macrophages of positively charged nanoparticles. This work provides improved diagnostic capability and a better understanding of the molecular imaging of atherosclerosis.
Collapse
Affiliation(s)
- Lingyi Wen
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Xiaomin Fu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Huan Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, China
- School of Medicine, Northwest University, Xi'an 710069, China
- Department of Radiology, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Pengfei Ye
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Hang Fu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Zhongqin Zhou
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Ran Sun
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Ting Xu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Chuan Fu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Chengcheng Zhu
- Department of Radiology, University of Washington, Seattle, Washington 98105, United States
| | - Yingkun Guo
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Haiming Fan
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, China
- School of Medicine, Northwest University, Xi'an 710069, China
| |
Collapse
|
12
|
Ryu JY, Jang EH, Lee J, Kim JH, Youn YN. Prevention of neointimal hyperplasia after coronary artery bypass graft via local delivery of sirolimus and rosuvastatin: network pharmacology and in vivo validation. J Transl Med 2024; 22:166. [PMID: 38365767 PMCID: PMC10874014 DOI: 10.1186/s12967-024-04875-8] [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: 09/14/2023] [Accepted: 01/08/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Coronary artery bypass graft (CABG) is generally used to treat complex coronary artery disease. Treatment success is affected by neointimal hyperplasia (NIH) of graft and anastomotic sites. Although sirolimus and rosuvastatin individually inhibit NIH progression, the efficacy of combination treatment remains unknown. METHODS We identified cross-targets associated with CABG, sirolimus, and rosuvastatin by using databases including DisGeNET and GeneCards. GO and KEGG pathway enrichment analyses were conducted using R studio, and target proteins were mapped in PPI networks using Metascape and Cytoscape. For in vivo validation, we established a balloon-injured rabbit model by inducing NIH and applied a localized perivascular drug delivery device containing sirolimus and rosuvastatin. The outcomes were evaluated at 1, 2, and 4 weeks post-surgery. RESULTS We identified 115 shared targets between sirolimus and CABG among databases, 23 between rosuvastatin and CABG, and 96 among all three. TNF, AKT1, and MMP9 were identified as shared targets. Network pharmacology predicted the stages of NIH progression and the corresponding signaling pathways linked to sirolimus (acute stage, IL6/STAT3 signaling) and rosuvastatin (chronic stage, Akt/MMP9 signaling). In vivo experiments demonstrated that the combination of sirolimus and rosuvastatin significantly suppressed NIH progression. This combination treatment also markedly decreased the expression of inflammation and Akt signaling pathway-related proteins, which was consistent with the predictions from network pharmacology analysis. CONCLUSIONS Sirolimus and rosuvastatin inhibited pro-inflammatory cytokine production during the acute stage and regulated Akt/mTOR/NF-κB/STAT3 signaling in the chronic stage of NIH progression. These potential synergistic mechanisms may optimize treatment strategies to improve long-term patency after CABG.
Collapse
Affiliation(s)
- Ji-Yeon Ryu
- Division of Cardiovascular Surgery, Department of Thoracic and Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Eui Hwa Jang
- Division of Cardiovascular Surgery, Department of Thoracic and Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - JiYong Lee
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, South Korea
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Jung-Hwan Kim
- Division of Cardiovascular Surgery, Department of Thoracic and Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Young-Nam Youn
- Division of Cardiovascular Surgery, Department of Thoracic and Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
13
|
Russu E, Arbanasi EM, Chirila TV, Muresan AV. Therapeutic strategies based on non-ionizing radiation to prevent venous neointimal hyperplasia: the relevance for stenosed arteriovenous fistula, and the role of vascular compliance. Front Cardiovasc Med 2024; 11:1356671. [PMID: 38374996 PMCID: PMC10875031 DOI: 10.3389/fcvm.2024.1356671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
We have reviewed the development and current status of therapies based on exposure to non-ionizing radiation (with a photon energy less than 10 eV) aimed at suppressing the venous neointimal hyperplasia, and consequentially at avoiding stenosis in arteriovenous grafts. Due to the drawbacks associated with the medical use of ionizing radiation, prominently the radiation-induced cardiovascular disease, the availability of procedures using non-ionizing radiation is becoming a noteworthy objective for the current research. Further, the focus of the review was the use of such procedures for improving the vascular access function and assuring the clinical success of arteriovenous fistulae in hemodialysis patients. Following a brief discussion of the physical principles underlying radiotherapy, the current methods based on non-ionizing radiation, either in use or under development, were described in detail. There are currently five such techniques, including photodynamic therapy (PDT), far-infrared therapy, photochemical tissue passivation (PTP), Alucent vascular scaffolding, and adventitial photocrosslinking. The last three are contingent on the mechanical stiffening achievable by the exogenous photochemical crosslinking of tissular collagen, a process that leads to the decrease of venous compliance. As there are conflicting opinions on the role of compliance mismatch between arterial and venous conduits in a graft, this aspect was also considered in our review.
Collapse
Affiliation(s)
- Eliza Russu
- Clinic of Vascular Surgery, Mures County Emergency Hospital, Targu Mures, Romania
- Department of Vascular Surgery, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
| | - Emil-Marian Arbanasi
- Clinic of Vascular Surgery, Mures County Emergency Hospital, Targu Mures, Romania
- Department of Vascular Surgery, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
- Centre for Advanced Medical and Pharmaceutical Research (CCAMF), George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
| | - Traian V. Chirila
- Centre for Advanced Medical and Pharmaceutical Research (CCAMF), George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
- Queensland Eye Institute, Woolloongabba, QLD, Australia
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD, Australia
- Australian Institute of Bioengineering and Nanotechnology (AIBN), University of Queensland, St Lucia, QLD, Australia
| | - Adrian V. Muresan
- Clinic of Vascular Surgery, Mures County Emergency Hospital, Targu Mures, Romania
- Department of Vascular Surgery, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
| |
Collapse
|
14
|
Starodubtseva I, Meshkova M, Zuikova A. Pathogenetic mechanisms of repeated adverse cardiovascular events development in patients with coronary heart disease: the role of chronic inflammation. Folia Med (Plovdiv) 2023; 65:863-870. [PMID: 38351773 DOI: 10.3897/folmed.65.e109433] [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: 07/12/2023] [Accepted: 08/03/2023] [Indexed: 02/16/2024] Open
Abstract
Stent restenosis is the most unfavorable complication of interventional treatment for coronary heart disease. We already know from various literature sources that the causes for stent restenosis in patients are both mechanical damage (partial opening, stent breakage, extended stented area, calcification, incomplete stent coverage of atherosclerotic plaque, weak radial stiffness of the stent metal frame, lack of stent drug coating), and the neointimal hyperplasia formation which is closely related to the de novo atherosclerosis development, being a predictor of the recurrent cardiovascular event.
Collapse
Affiliation(s)
| | - Maria Meshkova
- NN Burdenko Voronezh State Medical University, Voronezh, Russia
| | - Anna Zuikova
- NN Burdenko Voronezh State Medical University, Voronezh, Russia
| |
Collapse
|
15
|
Tan RP, Hung JC, Chan AHP, Grant AJ, Moore MJ, Lam YT, Michael P, Wise SG. Highly reproducible rat arterial injury model of neointimal hyperplasia. PLoS One 2023; 18:e0290342. [PMID: 37590291 PMCID: PMC10434902 DOI: 10.1371/journal.pone.0290342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023] Open
Abstract
Models of arterial injury in rodents have been invaluable to our current understanding of vessel restenosis and play a continuing role in the development of endovascular interventions for cardiovascular disease. Mechanical distention of the vessel wall and denudation of the vessel endothelium are the two major modes of vessel injury observed in most clinical pathologies and are critical to the reproducible modelling of progressive neointimal hyperplasia. The current models which have dominated this research area are the mouse wire carotid or femoral injury and the rat carotid balloon injury. While these elicit simultaneous distension of the vessel wall and denudation of the luminal endothelium, each model carries limitations that need to be addressed using a complementary injury model. Wire injuries in mice are highly technical and procedurally challenging due to small vessel diameters, while rat balloon injuries require permanent blood vessel ligation and disruption of native blood flow. Complementary models of vascular injury with reproducibility, convenience, and increased physiological relevance to the pathophysiology of endovascular injury would allow for improved studies of neointimal hyperplasia in both basic and translational research. In this study, we developed a new surgical model that elicits vessel distention and endothelial denudation injury using sequential steps using microforceps and a standard needle catheter inserted via arteriotomy into a rat common carotid artery, without requiring permanent ligation of branching arteries. After 2 weeks post-injury this model elicits highly reproducible neointimal hyperplasia and rates of re-endothelialisation similar to current wire and balloon injury models. Furthermore, evaluation of the smooth muscle cell phenotype profile, inflammatory response and extracellular matrix within the developing neointima, showed that our model replicated the vessel remodelling outcomes critical to restenosis and those becoming increasingly focused upon in the development of new anti-restenosis therapies.
Collapse
Affiliation(s)
- Richard P. Tan
- Faculty of Health and Medicine, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Jui Chien Hung
- Faculty of Health and Medicine, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Alex H. P. Chan
- Faculty of Health and Medicine, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Angus J. Grant
- Faculty of Health and Medicine, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Matthew J. Moore
- Faculty of Health and Medicine, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Yuen Ting Lam
- Faculty of Health and Medicine, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Praveesuda Michael
- Faculty of Health and Medicine, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Steven G. Wise
- Faculty of Health and Medicine, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
16
|
Mechelinck M, Hein M, Kupp C, Braunschweig T, Helmedag MJ, Klinkenberg A, Habigt MA, Klinge U, Tolba RH, Uhlig M. Experimental Liver Cirrhosis Inhibits Restenosis after Balloon Angioplasty. Int J Mol Sci 2023; 24:11351. [PMID: 37511114 PMCID: PMC10379020 DOI: 10.3390/ijms241411351] [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: 06/19/2023] [Revised: 07/09/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The effect of liver cirrhosis on vascular remodeling in vivo remains unknown. Therefore, this study investigates the influence of cholestatic liver cirrhosis on carotid arterial remodeling. A total of 79 male Sprague Dawley rats underwent bile duct ligation (cirrhotic group) or sham surgery (control group) and 28 days later left carotid artery balloon dilatation; 3, 7, 14 and 28 days after balloon dilatation, the rats were euthanized and carotid arteries were harvested. Histological sections were planimetrized, cell counts determined, and systemic inflammatory parameters measured. Up to day 14 after balloon dilatation, both groups showed a comparable increase in neointima area and degree of stenosis. By day 28, however, both values were significantly lower in the cirrhotic group (% stenosis: 20 ± 8 vs. 42 ± 10, p = 0.010; neointimal area [mm2]: 0.064 ± 0.025 vs. 0.138 ± 0.025, p = 0.024). Simultaneously, cell density in the neointima (p = 0.034) and inflammatory parameters were significantly higher in cirrhotic rats. This study demonstrates that cholestatic liver cirrhosis in rats substantially increases neointimal cell consolidation between days 14 and 28. Thereby, consolidation proved important for the degree of stenosis. This may suggest that patients with cholestatic cirrhosis are at lower risk for restenosis after coronary intervention.
Collapse
Affiliation(s)
- Mare Mechelinck
- Department of Anesthesiology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Marc Hein
- Department of Anesthesiology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Carolin Kupp
- Department of Anesthesiology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Till Braunschweig
- Department of Pathology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Marius J Helmedag
- Department of General, Visceral and Transplantation Surgery, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Axel Klinkenberg
- Department of Anesthesiology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Moriz A Habigt
- Department of Anesthesiology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Uwe Klinge
- Department of General, Visceral and Transplantation Surgery, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - René H Tolba
- Institute for Laboratory Animal Science and Experimental Surgery, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Moritz Uhlig
- Department of Anesthesiology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
17
|
Wu M, Xun M, Chen Y. Adaptation of Vascular Smooth Muscle Cell to Degradable Metal Stent Implantation. ACS Biomater Sci Eng 2023. [PMID: 37364226 DOI: 10.1021/acsbiomaterials.3c00637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Iron-, magnesium-, or zinc-based metal vessel stents support vessel expansion at the period early after implantation and degrade away after vascular reconstruction, eliminating the side effects due to the long stay of stent implants in the body and the risks of restenosis and neoatherosclerosis. However, emerging evidence has indicated that their degradation alters the vascular microenvironment and induces adaptive responses of surrounding vessel cells, especially vascular smooth muscle cells (VSMCs). VSMCs are highly flexible cells that actively alter their phenotype in response to the stenting, similarly to what they do during all stages of atherosclerosis pathology, which significantly influences stent performance. This Review discusses how biodegradable metal stents modify vascular conditions and how VSMCs respond to various chemical, biological, and physical signals attributable to stent implantation. The focus is placed on the phenotypic adaptation of VSMCs and the clinical complications, which highlight the importance of VSMC transformation in future stent design.
Collapse
Affiliation(s)
- Meichun Wu
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
- School of Nursing, University of South China, Hengyang, Hunan 410001, China
| | - Min Xun
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang, Hunan 410001, China
| | - Yuping Chen
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang, Hunan 410001, China
| |
Collapse
|
18
|
Govatati S, Pichavaram P, Kumar R, Rao GN. Blockade of CD47 function attenuates restenosis by promoting smooth muscle cell efferocytosis and inhibiting their migration and proliferation. J Biol Chem 2023; 299:104594. [PMID: 36898577 PMCID: PMC10124914 DOI: 10.1016/j.jbc.2023.104594] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 03/10/2023] Open
Abstract
Cluster of differentiation 47 (CD47) plays an important role in the pathophysiology of various diseases including atherosclerosis, but its role in neointimal hyperplasia which contributes to restenosis, has not been studied. Using molecular approaches in combination with a mouse vascular endothelial denudation model, we studied the role of CD47 in injury-induced neointimal hyperplasia. We determined that thrombin induced CD47 expression both in human and mouse aortic smooth muscle cells (HASMCs and MASMCs). In exploring the mechanisms, we found that the protease-activated receptor 1 (PAR1)-Gα protein q/11 (Gαq/11)-phospholipase Cβ3 (PLCβ3)-nuclear factor of activated T cells c1 (NFATc1) signaling axis regulates thrombin-induced CD47 expression in HASMCs. Depletion of CD47 levels using its siRNA or interference of its function by its blocking antibody (bAb) blunted thrombin-induced migration and proliferation of HASMCs and MASMCs. In addition, we found that thrombin-induced HASMC migration requires CD47 interaction with integrin β3. On the other hand, thrombin-induced HASMC proliferation was dependent on CD47's role in nuclear export and degradation of CDK-interacting protein 1 (p21Cip1). In addition, suppression of CD47 function by its bAb rescued HASMC efferocytosis from inhibition by thrombin. We also found that vascular injury induces CD47 expression in intimal SMCs and that inhibition of CD47 function by its bAb, while alleviating injury-induced inhibition of SMC efferocytosis, attenuated SMC migration and proliferation resulting in reduced neointima formation. Thus, these findings reveal a pathological role for CD47 in neointimal hyperplasia.
Collapse
Affiliation(s)
- Suresh Govatati
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Prahalathan Pichavaram
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Raj Kumar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Gadiparthi N Rao
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
19
|
Warwick T, Buchmann GK, Pflüger-Müller B, Spaeth M, Schürmann C, Abplanalp W, Tombor L, John D, Weigert A, Leo-Hansmann M, Dimmeler S, Brandes RP. Acute injury to the mouse carotid artery provokes a distinct healing response. Front Physiol 2023; 14:1125864. [PMID: 36824462 PMCID: PMC9941170 DOI: 10.3389/fphys.2023.1125864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/23/2023] [Indexed: 02/10/2023] Open
Abstract
Treatment of vascular stenosis with angioplasty results in acute vascular damage, which may lead to restenosis. Owing to the highly complex cellularity of blood vessels, the healing response following this damage is incompletely understood. To gain further insight into this process, scRNA-seq of mouse carotid tissue after wire injury was performed. Stages of acute inflammation, resolution and remodeling were recapitulated in these data. To identify cell types which give rise to neointima, analyses focused on smooth muscle cell and fibroblast populations, and included data integration with scRNA-seq data from myocardial infarction and atherosclerosis datasets. Following carotid injury, a subpopulation of smooth muscle cells which also arises during atherosclerosis and myocardial infarction was identified. So-called stem cell/endothelial cell/monocyte (SEM) cells are candidates for repopulating injured vessels, and were amongst the most proliferative cell clusters following wire-injury of the carotid artery. Importantly, SEM cells exhibit specific transcriptional profiles which could be therapeutically targeted. SEM cell gene expression patterns could also be detected in bulk RNA-sequencing of neointimal tissue isolated from injured carotid vessels by laser capture microdissection. These data indicate that phenotypic plasticity of smooth muscle cells is highly important to the progression of lumen loss following acute carotid injury. Interference with SEM cell formation could be an innovative approach to combat development of restenosis.
Collapse
Affiliation(s)
- Timothy Warwick
- Institute for Cardiovascular Physiology, Goethe University Frankfurt, Frankfurt am Main, Germany,German Center for Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany
| | - Giulia Karolin Buchmann
- Institute for Cardiovascular Physiology, Goethe University Frankfurt, Frankfurt am Main, Germany,German Center for Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany
| | - Beatrice Pflüger-Müller
- Institute for Cardiovascular Physiology, Goethe University Frankfurt, Frankfurt am Main, Germany,German Center for Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany
| | - Manuela Spaeth
- Institute for Cardiovascular Physiology, Goethe University Frankfurt, Frankfurt am Main, Germany,German Center for Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany
| | - Christoph Schürmann
- Institute for Cardiovascular Physiology, Goethe University Frankfurt, Frankfurt am Main, Germany,German Center for Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany
| | - Wesley Abplanalp
- German Center for Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany,Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Lukas Tombor
- German Center for Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany,Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - David John
- German Center for Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany,Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Martin Leo-Hansmann
- Department of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Stefanie Dimmeler
- German Center for Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany,Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Ralf P. Brandes
- Institute for Cardiovascular Physiology, Goethe University Frankfurt, Frankfurt am Main, Germany,German Center for Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany,*Correspondence: Ralf P. Brandes,
| |
Collapse
|
20
|
Chen J, Zhang H, Li L, Zhang X, Zhao D, Wang L, Wang J, Yang P, Sun H, Liu K, Chen W, Li L, Lin F, Li Z, Chen YE, Zhang J, Pang D, Ouyang H, He Y, Fan J, Tang X. Lp-PLA 2 (Lipoprotein-Associated Phospholipase A 2) Deficiency Lowers Cholesterol Levels and Protects Against Atherosclerosis in Rabbits. Arterioscler Thromb Vasc Biol 2023; 43:e11-e28. [PMID: 36412196 DOI: 10.1161/atvbaha.122.317898] [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] [Indexed: 11/23/2022]
Abstract
BACKGROUND Elevated plasma Lp-PLA2 (lipoprotein-associated phospholipase A2) activity is closely associated with an increased risk of cardiovascular events. However, whether and how Lp-PLA2 is directly involved in the pathogenesis of atherosclerosis is still unclear. To examine the hypothesis that Lp-PLA2 could be a potential preventative target of atherosclerosis, we generated Lp-PLA2 knockout rabbits and investigated the pathophysiological functions of Lp-PLA2. METHODS Lp-PLA2 knockout rabbits were generated using CRISPR/Cas9 system to assess the role of Lp-PLA2 in plasma lipids regulation and identify its underlying molecular mechanisms. Homozygous knockout rabbits along with wild-type rabbits were fed a cholesterol-rich diet for up to 14 weeks and their atherosclerotic lesions were compared. Moreover, the effects of Lp-PLA2 deficiency on the key cellular behaviors in atherosclerosis were assessed in vitro. RESULTS When rabbits were fed a standard diet, Lp-PLA2 deficiency led to a significant reduction in plasma lipids. The decreased protein levels of SREBP2 (sterol regulatory element-binding protein 2) and HMGCR (3-hydroxy-3-methylglutaryl coenzyme A reductase) in livers of homozygous knockout rabbits indicated that the cholesterol biosynthetic pathway was impaired with Lp-PLA2 deficiency. In vitro experiments further demonstrated that intracellular Lp-PLA2 efficiently enhanced SREBP2-related cholesterol biosynthesis signaling independently of INSIGs (insulin-induced genes). When fed a cholesterol-rich diet, homozygous knockout rabbits exhibited consistently lower level of hypercholesterolemia, and their aortic atherosclerosis lesions were significantly reduced by 60.2% compared with those of wild-type rabbits. The lesions of homozygous knockout rabbits were characterized by reduced macrophages and the expression of inflammatory cytokines. Macrophages of homozygous knockout rabbits were insensitive to M1 polarization and showed reduced DiI-labeled lipoprotein uptake capacity compared with wild-type macrophages. Lp-PLA2 deficiency also inhibited the adhesion between monocytes and endothelial cells. CONCLUSIONS These results demonstrate that Lp-PLA2 plays a causal role in regulating blood lipid homeostasis and Lp-PLA2 deficiency protects against dietary cholesterol-induced atherosclerosis in rabbits. Lp-PLA2 could be a potential target for the prevention of atherosclerosis.
Collapse
Affiliation(s)
- Jiahuan Chen
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Huanyu Zhang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Linquan Li
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Xinwei Zhang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Dazhong Zhao
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Lingyu Wang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Jiaqi Wang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Ping Yang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China (P.Y., H.S., K.L., W.C., Y.H.)
| | - Huan Sun
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China (P.Y., H.S., K.L., W.C., Y.H.)
| | - Kun Liu
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China (P.Y., H.S., K.L., W.C., Y.H.)
| | - Weiwei Chen
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China (P.Y., H.S., K.L., W.C., Y.H.)
| | - Lin Li
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Feng Lin
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Zhanjun Li
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Y Eugene Chen
- Department of Internal Medicine, Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor (Y.E.C., J.Z.)
| | - Jifeng Zhang
- Department of Internal Medicine, Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor (Y.E.C., J.Z.)
| | - Daxin Pang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.).,Chongqing Research Institute, Jilin University, Chongqing, China (D.P., H.O., X.T.)
| | - Hongsheng Ouyang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.).,Chongqing Research Institute, Jilin University, Chongqing, China (D.P., H.O., X.T.)
| | - Yuquan He
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China (P.Y., H.S., K.L., W.C., Y.H.)
| | - Jianglin Fan
- Department of Molecular Pathology, Faculty of Medicine, Graduate School of Medical Sciences, University of Yamanashi, Japan (J.F.)
| | - Xiaochun Tang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.).,Chongqing Research Institute, Jilin University, Chongqing, China (D.P., H.O., X.T.)
| |
Collapse
|
21
|
Sareło P, Sobieszczańska B, Wysokińska E, Gąsior-Głogowska M, Kałas W, Podbielska H, Wawrzyńska M, Kopaczyńska M. In vitro examinations of the anti-inflammatory interleukin functionalized polydopamine based biomaterial as a potential coating for cardiovascular stents. Biocybern Biomed Eng 2023. [DOI: 10.1016/j.bbe.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
|
22
|
Bhargava A, Sandoval Castellanos AM, Shah S, Ning K. An insight into the iPSCs-derived two-dimensional culture and three-dimensional organoid models for neurodegenerative disorders. Interface Focus 2022; 12:20220040. [PMID: 35992771 PMCID: PMC9372641 DOI: 10.1098/rsfs.2022.0040] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/18/2022] [Indexed: 12/20/2022] Open
Abstract
The use of induced pluripotent stem cells (iPSCs) is a promising approach when used as models to study neurodegenerative disorders (NDDs) in vitro. iPSCs have been used in in vitro two-dimensional cultures; however, these two-dimensional cultures do not mimic the physiological three-dimensional cellular environment. The use of iPSCs-derived three-dimensional organoids has risen as a powerful alternative to using animal models to study NDDs. These iPSCs-derived three-dimensional organoids can resemble the complexity of the tissue of interest, making it an approachable, cost-effective technique, to study NDDs in an ethical manner. Furthermore, the use of iPSCs-derived organoids will be an important tool to develop new therapeutics and pharmaceutics to treat NDDs. Herein, we will highlight how iPSCs-derived two-dimensional cultures and three-dimensional organoids have been used to study NDDs, as well as the advantages and disadvantages of both techniques.
Collapse
Affiliation(s)
- Anushka Bhargava
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, The University of Sheffield, Sheffield S10 2HQ, UK
| | - Ana M. Sandoval Castellanos
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, The University of Sheffield, Sheffield S10 2HQ, UK
| | - Sonali Shah
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, The University of Sheffield, Sheffield S10 2HQ, UK
| | - Ke Ning
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, The University of Sheffield, Sheffield S10 2HQ, UK
| |
Collapse
|
23
|
Mukhamadiyarov RA, Koshelev VA, Frolov AV, Mironov AV, Shabaev AR, Evtushenko AV, Lyapin AA, Kutikhin AG. [Ultrastructure of neointima of native and artificial elements of the blood circulatory system]. Arkh Patol 2022; 84:14-23. [PMID: 35639839 DOI: 10.17116/patol20228403114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To compare the neointima structure in conduits for coronary bypass grafting, bioprosthetic heart valves, tissue-engineered vascular grafts, and metal stents. MATERIAL AND METHODS The objects of the study were the fragments of the human internal thoracic artery, experimental biodegradable vascular prostheses, leaflets of xenopericardial bioprostheses of heart valves, and fragments of stented vessels. Tissue samples were fixed in formalin and post-fixed in osmium tetroxide. After dehydration and epoxy resin embedding, the samples were ground and polished. Samples were counterstained with uranyl acetate and lead citrate and visualized by means of backscattered scanning electron microscopy. RESULTS Neointimal pattern in all samples was similar. Neointima was comprised of endothelial cells, smooth muscle cells, fibroblasts, and the extracellular matrix. Endothelial cells showed significant diversity both between different elements of the circulatory system and within the same tissue, having either elongated or polygonal shape. Adhesion of leukocytes testified to the endothelial cell activation. In the absence of inflammation in the superficial layer of the neointima, the arrangement of smooth muscle cells and extracellular matrix fibers was parallel to the endothelium. Clusters of foam cells were frequently detected around the neointimal layers with solid inclusions (metal stents or calcium deposits). Thickening of the neointima was accompanied by the presence of capillaries and capillary-like structures. CONCLUSION Neointima formation is a typical response to the damage inflicted to the elements of the circulatory system. Neointima underwent a constant remodeling characterized by an altered cellular composition, macrophage invasion, neovascularization, and calcification.
Collapse
Affiliation(s)
- R A Mukhamadiyarov
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - V A Koshelev
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - A V Frolov
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - A V Mironov
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - A R Shabaev
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - A V Evtushenko
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - A A Lyapin
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - A G Kutikhin
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| |
Collapse
|
24
|
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: 2.3] [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
|
25
|
Kanaan R, Medlej-Hashim M, Jounblat R, Pilecki B, Sorensen GL. Microfibrillar-associated protein 4 in health and disease. Matrix Biol 2022; 111:1-25. [DOI: 10.1016/j.matbio.2022.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/04/2022] [Accepted: 05/24/2022] [Indexed: 10/18/2022]
|