1
|
Won DS, Lee KB, Park JH, Zeng CH, Kim MH, Lee DH. Balloon neck-plasty to create a wide-necked aneurysm in the elastase-induced rabbit model. Neuroradiology 2024; 66:825-834. [PMID: 38438630 DOI: 10.1007/s00234-024-03326-x] [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: 08/07/2023] [Accepted: 02/24/2024] [Indexed: 03/06/2024]
Abstract
PURPOSE The elastase-induced aneurysm (EIA) model in rabbits has been proposed for translational research; however, the adjustment of aneurysm neck size remains challenging. In this study, the technical feasibility and safety of balloon neck-plasty to create a wide-necked aneurysm in rabbit EIA model were investigated. METHODS Male New Zealand White rabbits (N = 15) were randomly assigned to three groups: group A, EIA creation without neck-plasty; group B, neck-plasty immediately after EIA creation; group C, neck-plasty 4 weeks after EIA creation. The diameter of balloon used for neck-plasty was determined 1 mm larger than origin carotid artery diameter. All rabbits were euthanized 4 weeks after their final surgery. Aneurysm neck, height, dome-to-neck (D/N) ratio, and histologic parameters were compared among the groups. RESULTS Aneurysm creation was technically successful in 14 out of 15 rabbits (93.3%), with one rabbit experiencing mortality due to an adverse anesthetic event during the surgery. Saccular and wide-necked aneurysms were successfully created in all rabbits. Aneurysm neck was significantly greater in groups B and C compared to group A (all P < .05). D/N ratio was significantly lower in groups B and C compared to group A (all P < .05). Additionally, tunica media thickness, vessel area, and luminal area were significantly greater in groups B and C compared to group A (all P < .05). These variables were found to be significantly greater in group B compared to group C (all P < .05). CONCLUSION The creation of a wide-necked aneurysm using balloon neck-plasty after elastase induction in rabbits has been determined to be technically feasible and safe.
Collapse
Affiliation(s)
- Dong-Sung Won
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
- Department of Radiology, Asan Medical Center, Research Institute of Radiology, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Ki Baek Lee
- Department of Radiologic Technology, Chungbuk Health and Science University, Cheongju, Republic of Korea
| | - Jung-Hoon Park
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Chu Hui Zeng
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
- Department of Radiology, Asan Medical Center, Research Institute of Radiology, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Mi Hyeon Kim
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
- Department of Radiology, Asan Medical Center, Research Institute of Radiology, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Deok Hee Lee
- Department of Radiology, Asan Medical Center, Research Institute of Radiology, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
2
|
Lin A, Brittan M, Baker AH, Dimmeler S, Fisher EA, Sluimer JC, Misra A. Clonal Expansion in Cardiovascular Pathology. JACC Basic Transl Sci 2024; 9:120-144. [PMID: 38362345 PMCID: PMC10864919 DOI: 10.1016/j.jacbts.2023.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 02/17/2024]
Abstract
Clonal expansion refers to the proliferation and selection of advantageous "clones" that are better suited for survival in a Darwinian manner. In recent years, we have greatly enhanced our understanding of cell clonality in the cardiovascular context. However, our knowledge of the underlying mechanisms behind this clonal selection is still severely limited. There is a transpiring pattern of clonal expansion of smooth muscle cells and endothelial cells-and, in some cases, macrophages-in numerous cardiovascular diseases irrespective of their differing microenvironments. These findings indirectly suggest the possible existence of stem-like vascular cells which are primed to respond during disease. Subsequent clones may undergo further phenotypic changes to adopt either protective or detrimental roles. By investigating these clone-forming vascular cells, we may be able to harness this inherent clonal nature for future therapeutic intervention. This review comprehensively discusses what is currently known about clonal expansion across the cardiovascular field. Comparisons of the clonal nature of vascular cells in atherosclerosis (including clonal hematopoiesis of indeterminate potential), pulmonary hypertension, aneurysm, blood vessel injury, ischemia- and tumor-induced angiogenesis, and cerebral cavernous malformations are evaluated. Finally, we discuss the potential clinical implications of these findings and propose that proper understanding and specific targeting of these clonal cells may provide unique therapeutic options for the treatment of these cardiovascular conditions.
Collapse
Affiliation(s)
- Alexander Lin
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, New South Wales, Australia
| | - Mairi Brittan
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew H. Baker
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- CARIM School for Cardiovascular Sciences, Department of Pathology, Maastricht University Medical Center (MUMC), Maastricht, the Netherlands
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Edward A. Fisher
- Department of Medicine/Division of Cardiology, New York University Grossman School of Medicine, New York, New York, USA
- Cardiovascular Research Center, New York University Grossman School of Medicine, New York, New York, USA
| | - Judith C. Sluimer
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- CARIM School for Cardiovascular Sciences, Department of Pathology, Maastricht University Medical Center (MUMC), Maastricht, the Netherlands
| | - Ashish Misra
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
3
|
Rusin CG, Acosta SI, Brady KM, Vu E, Scahill C, Fonseca B, Barrett C, Simsic J, Yates AR, Klepczynski B, Gaynor WJ, Penny DJ. Automated prediction of cardiorespiratory deterioration in patients with single-ventricle parallel circulation: A multicenter validation study. JTCVS OPEN 2023; 15:406-411. [PMID: 37808061 PMCID: PMC10556807 DOI: 10.1016/j.xjon.2023.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/13/2023] [Accepted: 05/02/2023] [Indexed: 10/10/2023]
Abstract
Objectives Patients with single-ventricle physiology have a significant risk of cardiorespiratory deterioration between their first- and second-stage palliation surgeries. Detection of deterioration episodes may allow for early intervention and improved outcomes. Methods A prospective study was executed at Nationwide Children's Hospital, Children's Hospital of Philadelphia, and Children's Hospital Colorado to collect physiologic data of subjects with single ventricle physiology during all hospitalizations between neonatal palliation and II surgeries using the Sickbay software platform (Medical Informatics Corp). Timing of cardiorespiratory deterioration events was captured via chart review. The predictive algorithm previously developed and validated at Texas Children's Hospital was applied to these data without retraining. Standard metrics such as receiver operating curve area, positive and negative likelihood ratio, and alert rates were calculated to establish clinical performance of the predictive algorithm. Results Our cohort consisted of 58 subjects admitted to the cardiac intensive care unit and stepdown units of participating centers over 14 months. Approximately 28,991 hours of high-resolution physiologic waveform and vital sign data were collected using the Sickbay. A total of 30 cardiorespiratory deterioration events were observed. the risk index metric generated by our algorithm was found to be both sensitive and specific for detecting impending events one to two hours in advance of overt extremis (receiver operating curve = 0.927). Conclusions Our algorithm can provide a 1- to 2-hour advanced warning for 53.6% of all cardiorespiratory deterioration events in children with single ventricle physiology during their initial postop course as well as interstage hospitalizations after stage I palliation with only 2.5 alarms being generated per patient per day.
Collapse
Affiliation(s)
- Craig G. Rusin
- Department of Pediatrics—Cardiology, Baylor College of Medicine, Texas Children's Hospital, Houston, Tex
| | - Sebastian I. Acosta
- Department of Pediatrics—Cardiology, Baylor College of Medicine, Texas Children's Hospital, Houston, Tex
| | - Kennith M. Brady
- Department of Anesthesiology, Northwestern University, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill
| | - Eric Vu
- Department of Anesthesiology, Northwestern University, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill
| | - Carly Scahill
- Department of Pediatrics—Cardiology, Children's Hospital Colorado, Aurora, Colo
| | - Brian Fonseca
- Department of Pediatrics—Cardiology, Children's Hospital Colorado, Aurora, Colo
| | - Cindy Barrett
- Department of Pediatrics—Cardiology, Children's Hospital Colorado, Aurora, Colo
| | - Janet Simsic
- Department of Pediatrics—Cardiology, Nationwide Children's Hospital, Columbus, Ohio
| | - Andrew R. Yates
- Department of Pediatrics—Cardiology, Nationwide Children's Hospital, Columbus, Ohio
| | - Brenna Klepczynski
- Department of Cardiovascular Surgery, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - William J. Gaynor
- Department of Cardiovascular Surgery, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Daniel J. Penny
- Department of Pediatrics—Cardiology, Baylor College of Medicine, Texas Children's Hospital, Houston, Tex
| |
Collapse
|
4
|
Sobolevskaya EV, Shumkov OA, Smagin MA, Guskov AE, Malysheva AV, Atuchin VV, Nimaev VV. Markers of Restenosis after Percutaneous Transluminal Balloon Angioplasty in Patients with Critical Limb Ischemia. Int J Mol Sci 2023; 24:ijms24109096. [PMID: 37240440 DOI: 10.3390/ijms24109096] [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: 03/30/2023] [Revised: 04/14/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Among cardiovascular diseases, chronic obliterating lesions of the arteries of lower extremities, which are one of the important problems of modern healthcare, are distinguished. In most cases, the cause of damage to the arteries of lower extremities is atherosclerosis. The most severe form is chronic ischemia, characterized by pain at rest and ischemic ulcers, ultimately increasing the risk of limb loss and cardiovascular mortality. Therefore, patients with critical limb ischemia need limb revascularization. Percutaneous transluminal balloon angioplasty is one of the least invasive and safe approaches, with advantages for patients with comorbidities. However, after this procedure, restenosis is still possible. Early detection of changes in the composition of some molecules as markers of restenosis will help screen patients at the risk of restenosis, as well as find ways to apply efforts for further directions of inhibition of this process. The purpose of this review is to provide the most important and up-to-date information on the mechanisms of restenosis development, as well as possible predictors of their occurrence. The information collected in this publication may be useful in predicting outcomes after surgical treatment and will also find new ways for the target implication to the mechanisms of development of restenosis and atherosclerosis.
Collapse
Affiliation(s)
- Elvira V Sobolevskaya
- Laboratory of Surgical Lymphology and Lymph-Detoxication, Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, SB RAS, Novosibirsk 630117, Russia
| | - Oleg A Shumkov
- Laboratory of Surgical Lymphology and Lymph-Detoxication, Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, SB RAS, Novosibirsk 630117, Russia
| | - Mikhail A Smagin
- Laboratory of Surgical Lymphology and Lymph-Detoxication, Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, SB RAS, Novosibirsk 630117, Russia
| | - Andrey E Guskov
- Laboratory of Scientometrics and Scientific Communications, Russian Research Institute of Economics, Politics and Law in Science and Technology, Moscow 127254, Russia
| | - Alexandra V Malysheva
- Laboratory of Scientometrics and Scientific Communications, Russian Research Institute of Economics, Politics and Law in Science and Technology, Moscow 127254, Russia
| | - Victor V Atuchin
- Laboratory of Optical Materials and Structures, Institute of Semiconductor Physics, SB RAS, Novosibirsk 630090, Russia
- Research and Development Department, Kemerovo State University, Kemerovo 650000, Russia
- Department of Industrial Machinery Design, Novosibirsk State Technical University, Novosibirsk 630073, Russia
- R&D Center "Advanced Electronic Technologies", Tomsk State University, Tomsk 634034, Russia
| | - Vadim V Nimaev
- Laboratory of Surgical Lymphology and Lymph-Detoxication, Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, SB RAS, Novosibirsk 630117, Russia
| |
Collapse
|
5
|
Rusin CG, Acosta SI, Vu EL, Ahmed M, Brady KM, Penny DJ. Automated Prediction of Cardiorespiratory Deterioration in Patients With Single Ventricle. J Am Coll Cardiol 2021; 77:3184-3192. [PMID: 34167643 DOI: 10.1016/j.jacc.2021.04.072] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/26/2021] [Accepted: 04/13/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Patients with single-ventricle physiology have a significant risk of cardiorespiratory deterioration between their first and second stage palliation surgeries. OBJECTIVES The objective of this study is to develop and validate a real-time computer algorithm that can automatically recognize physiological precursors of cardiorespiratory deterioration in children with single-ventricle physiology during their interstage hospitalization. METHODS A retrospective study was conducted from prospectively collected physiological data of subjects with single-ventricle physiology. Deterioration events were defined as a cardiac arrest requiring cardiopulmonary resuscitation or an unplanned intubation. Physiological metrics were derived from the electrocardiogram (heart rate, heart rate variability, ST-segment elevation, and ST-segment variability) and the photoplethysmogram (peripheral oxygen saturation and pleth variability index). A logistic regression model was trained to separate the physiological dynamics of the pre-deterioration phase from all other data generated by study subjects. Data were split 50/50 into model training and validation sets to enable independent model validation. RESULTS Our cohort consisted of 238 subjects admitted to the cardiac intensive care unit and stepdown units of Texas Children's Hospital over a period of 6 years. Approximately 300,000 h of high-resolution physiological waveform and vital sign data were collected using the Sickbay software platform (Medical Informatics Corp., Houston, Texas). A total of 112 cardiorespiratory deterioration events were observed. Seventy-two of the subjects experienced at least 1 deterioration event. The risk index metric generated by our optimized algorithm was found to be both sensitive and specific for detecting impending events 1 to 2 h in advance of overt extremis (receiver-operating characteristic curve area: 0.958; 95% confidence interval: 0.950 to 0.965). CONCLUSIONS Our algorithm can provide 1 to 2 h of advanced warning for 62% of all cardiorespiratory deterioration events in children with single-ventricle physiology during their interstage period, with only 1 alarm being generated at the bedside per patient per day.
Collapse
Affiliation(s)
- Craig G Rusin
- Department of Pediatrics-Cardiology, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA.
| | - Sebastian I Acosta
- Department of Pediatrics-Cardiology, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Eric L Vu
- Department of Anesthesiology, Northwestern University, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois, USA
| | - Mubbasheer Ahmed
- Department of Pediatrics-Critical Care, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Kennith M Brady
- Department of Anesthesiology, Northwestern University, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois, USA
| | - Daniel J Penny
- Department of Pediatrics-Cardiology, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
6
|
Satish M, Gunasekar P, Asensio JA, Agrawal DK. Vitamin D attenuates HMGB1-mediated neointimal hyperplasia after percutaneous coronary intervention in swine. Mol Cell Biochem 2020; 474:219-228. [PMID: 32737774 DOI: 10.1007/s11010-020-03847-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/20/2020] [Indexed: 12/15/2022]
Abstract
Intracoronary stenting is a common procedure in patients with coronary artery disease (CAD). Stent deployment stretches and denudes the endothelial layer, promoting a local inflammatory response, resulting in neointimal hyperplasia. Vitamin D deficiency associates with CAD. In this study, we examined the association of vitamin D status with high mobility group box 1 (HMGB1)-mediated pathways (HMGB1, receptor for advanced glycation end products [RAGE], and Toll-like receptor-2 and -4 [TLR2 and TLR4]) in neointimal hyperplasia in atherosclerotic swine following bare metal stenting. Yucatan microswine fed with a high-cholesterol diet were stratified to receive vitamin D-deficient (VD-DEF), vitamin D-sufficient (VD-SUF), and vitamin D-supplemented (VD-SUP) diet. After 6 months, PTCA (percutaneous transluminal balloon angioplasty) followed by bare metal stent implantation was performed in the left anterior descending (LAD) artery of each swine. Four months following coronary intervention, angiogram and optical coherence tomography (OCT) were performed and swine euthanized. Histology and immunohistochemistry were performed in excised LAD to evaluate the expression of HMGB1, RAGE, TLR2, and TLR4. OCT analysis revealed the greatest in-stent restenosis area in the LAD of VD-DEF compared to VD-SUF or VD-SUP swine. The protein expression of HMGB1, RAGE, TLR2, and TLR4 was significantly higher in the LAD of VD-DEF compared to VD-SUF or VD-SUP swine. Vitamin D deficiency was associated with both increased in-stent restenosis and increased HMGB1-mediated inflammation noted in coronary arteries following intravascular stenting. Inversely, vitamin D supplementation was associated with both a decrease in this inflammatory profile and in neointimal hyperplasia, warranting further investigation for vitamin D as a potential adjunct therapy following coronary intervention.
Collapse
Affiliation(s)
- Mohan Satish
- Department of Clinical & Translational Science, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE, 68178, USA
| | - Palanikumar Gunasekar
- Department of Clinical & Translational Science, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE, 68178, USA
| | - Juan A Asensio
- Department of Clinical & Translational Science, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE, 68178, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA.
| |
Collapse
|
7
|
Zhou C, Feng X, Shi Z, Song C, Cui X, Zhang J, Li T, Toft ES, Ge J, Wang L, Zhang H. Research on elastic recoil and restoration of vessel pulsatility of Zn-Cu biodegradable coronary stents. ACTA ACUST UNITED AC 2020; 65:219-227. [PMID: 31527289 DOI: 10.1515/bmt-2019-0025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 06/24/2019] [Indexed: 11/15/2022]
Abstract
Coronary stents made of zinc (Zn)-0.8 copper (Cu) (in wt%) alloy were developed as biodegradable metal stents (Zn-Cu stents) in this study. The mechanical properties of the Zn-Cu stents and the possible gain effects were characterized by in vitro and in vivo experiments compared with 316L stainless steel stents (316L stents). Young's modulus of the as-extruded Zn-0.8Cu alloy and properties of the stents, including their intrinsic elastic recoil, stent trackability were evaluated compared with 316L stents. In vivo study was also conducted to evaluate restoration of pulsatility of vessel segment implanted stents. Both Zn-Cu stents and 316L stents have good acute lumen gain. By comparison, the advantages of Zn-Cu stents are as follows: (I) Zn-Cu stents have less intrinsic elastic recoil than 316L stents; (II) stent trackability indicates that Zn-Cu stents have a smaller push force when passing through curved blood vessels, which may cause less mechanical stimulation to blood vessels; (III) in vivo study suggests that Zn-Cu stents implantation better facilitates the recovery of vascular pulsatility.
Collapse
Affiliation(s)
- Chao Zhou
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, P.R. China
| | - Xiangyi Feng
- National United Engineering Laboratory for Biomedical Material Modification, Branden Industrial Park, Qihe Economic and Development Zone, Dezhou City, Shandong 251100, P.R. China
| | - Zhangzhi Shi
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, P.R. China
| | - Caixia Song
- National United Engineering Laboratory for Biomedical Material Modification, Branden Industrial Park, Qihe Economic and Development Zone, Dezhou City, Shandong 251100, P.R. China
| | - Xiaoshan Cui
- National United Engineering Laboratory for Biomedical Material Modification, Branden Industrial Park, Qihe Economic and Development Zone, Dezhou City, Shandong 251100, P.R. China
| | - Junwei Zhang
- National United Engineering Laboratory for Biomedical Material Modification, Branden Industrial Park, Qihe Economic and Development Zone, Dezhou City, Shandong 251100, P.R. China
| | - Ting Li
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, P.R. China
| | - Egon Steen Toft
- Vice President for Medical and Health Sciences, Medical and Health Sciences Office, College of Medicine, Qatar University, Doha, Qatar
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, P.R. China
| | - Luning Wang
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, P.R. China
| | - Haijun Zhang
- Department of Interventional and Vascular Surgery, The Tenth People's Hospital of Shanghai, Tongji University, Shanghai 200072, P.R. China.,Branden Industrial Park, Qihe Economic and Development Zone, Dezhou City, Shandong 251100, P.R. China.,Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
8
|
Abundes-Velasco A, Jiménez-Rodríguez GM, Arias-Sánchez E, Damas-De Los Santos F, Martínez-Ríos MA, Molina-Méndez FJ, Sánchez-Pérez E, Arai-Ito Marco M, Rodríguez-Barriga E, Sánchez-Jara M, Aceves-Díaz González S, Rodríguez-Parra DA, Aranda-Fraustro A, Romero-Ibarra JL, Peña-Duque MA. Histological and Mechanical Behavior of INC 01 and 02 Bare Metal Stents Against a Commercial Stent: A Preclinical Study in a Porcine Model. Arch Med Res 2020; 51:406-412. [PMID: 32376040 DOI: 10.1016/j.arcmed.2020.03.016] [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: 07/25/2019] [Revised: 02/01/2020] [Accepted: 03/17/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND PCI is an expensive procedure in our population and it implies a huge cost for the institutions and National Health Service. AIM OF THE STUDY The main objective was to evaluate the technical and biological success of two stents designed in Mexico. METHODS Ten York pigs, 4-6 months of age, underwent implantation of the bare metal INC-01 (10 stents) and INC-02 (6 stents) coronary stent in addition to a conventional commercial stent (10 stents). Technical success was evaluated immediately with angiography and Intravascular Ultrasound IVUS, continued by a mean follow-up of 4 month and a final angiographic, IVUS and histological evaluation. RESULTS Initial technical success, angiography and IVUS between the three stents were not significant. One stent presented restenosis in follow-up (commercial stent), but all other stents presented excellent clinical outcome, satisfactory angiographic and IVUS results. Inflammation, proliferation and endothelialization between the stents had no major differences in histological analysis in a mean of 4 months follow-up. CONCLUSIONS In this pig model, the INC 01 and INC 02 stents showed the same delivering technical success, angiographic and IVUS features, biological and histological response compared to commercial last generation stents.
Collapse
Affiliation(s)
- Arturo Abundes-Velasco
- Subdirección de Proyectos de Innovación y Desarrollo Tecnológico, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | | | - Eduardo Arias-Sánchez
- Departamento de Hemodinámica, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - Félix Damas-De Los Santos
- Departamento de Hemodinámica, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | | | - Francisco Javier Molina-Méndez
- Departamento de Anestesiología Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - Efraín Sánchez-Pérez
- Subdirección de Innovación y Desarrollo Tecnológico, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - Minoru Arai-Ito Marco
- Subdirección de Innovación y Desarrollo Tecnológico, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - Erika Rodríguez-Barriga
- Departamento de Hemodinámica, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - Mauricio Sánchez-Jara
- Departamento de Hemodinámica, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - Sebastián Aceves-Díaz González
- Subdirección de Innovación y Desarrollo Tecnológico, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - David Alexis Rodríguez-Parra
- Subdirección de Innovación y Desarrollo Tecnológico, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - Alberto Aranda-Fraustro
- Departamento de Patología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - José Luis Romero-Ibarra
- Departamento de Hemodinámica, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - Marco Antonio Peña-Duque
- Subdirección de Innovación y Desarrollo Tecnológico, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México.
| |
Collapse
|
9
|
He F, Yu C, Liu T, Jia H. Ginsenoside Rg1 as an Effective Regulator of Mesenchymal Stem Cells. Front Pharmacol 2020; 10:1565. [PMID: 32038244 PMCID: PMC6989539 DOI: 10.3389/fphar.2019.01565] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 12/03/2019] [Indexed: 12/16/2022] Open
Abstract
Recently, breakthroughs have been made in the use of mesenchymal stem cells (MSCs) to treat various diseases. Several stem cell types have been authorized as drugs by the European Medicines Agency and the U.S. Food and Drug Administration. The Chinese official document “Notification of the management of stem cell clinical research (trial)” was also published in August 2015. Currently, China has approved 106 official stem cell clinical research filing agencies and 62 clinical research projects, which are mostly focused on MSC therapy. Hence, the optimization and development of stem cell drugs is imperative. During this process, maximizing MSC expansion, minimizing cell loss during MSC transplantation, improving the homing rate, precisely regulating the differentiation of MSCs, and reducing MSC senescence and apoptosis are major issues in MSC preclinical research. Similar to artemisinin extracted from the stems and leaves of Artemisia annua, ginsenoside Rg1 (Rg1) is purified from the root or stem of ginseng. In the human body, Rg1 regulates organ function, which is inseparable from its regulation of adult stem cells. Rg1 treatment may effectively regulate the proliferation, differentiation, senescence, and apoptosis of MSCs in different microenvironments in vitro or in vivo. In this review, we discuss recent advances in understanding the effect of Rg1 on MSCs and describe the issues that must be addressed and prospects regarding Rg1 regulation of MSCs in preclinical or clinical studies.
Collapse
Affiliation(s)
- Fang He
- Key Laboratory of Cell Engineering of Guizhou Province, The Affiliated Hospital of Zunyi Medical University, Zunyi, China.,Department of Oral and Maxillofacial Surgery, University Hospital of Tübingen, Tübingen, Germany
| | - Changyin Yu
- Department of Neurology, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Tao Liu
- Department of Neurology, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Huilin Jia
- School of Stomatology, Xi'an Medical University, Xi'an, China
| |
Collapse
|
10
|
Ishikawa O, Tanaka M, Konno K, Hasebe T, Horikawa A, Iijima A, Saito N, Takahashi K. Swine model of in-stent stenosis in the iliac artery evaluating the serial time course. Exp Anim 2018; 67:501-508. [PMID: 30068792 PMCID: PMC6219888 DOI: 10.1538/expanim.18-0027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
The aim of this study was to propose a new animal model evaluating the serial time course
of in-stent stenosis by repeated carotid artery catheterization in the same animal. 16
bare-metal stents were implanted in the normal external and internal iliac artery of 8
miniature pigs. Repeated measurements were performed in the same animal every 2 weeks for
12 weeks through carotid artery catheterization. The time course and peak neointimal
proliferation were evaluated by intravascular ultrasound. Health of all animals was
assessed by clinical and hematological examinations. As a result, 7 times of carotid
artery catheterization was performed per pig, but all animals remained healthy without
both any complications and hematological inflammatory abnormalities. The time course of
neointimal proliferation of each stent was observed from the stage of hyperplasia to
partial regression. The peak neointimal proliferation varied from 6 to 12 weeks despite
implantation of identical stents using the same deployment method. In conclusion, repeated
carotid artery catheterization to the same animal is feasible without animal health
deterioration. This model should be useful to evaluate the time course of neointimal
proliferation after stent deployment in preclinical study.
Collapse
Affiliation(s)
- Osamu Ishikawa
- Department of Neurosurgery, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Minoru Tanaka
- Department of Transfusion Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kenjiro Konno
- Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Terumitsu Hasebe
- Department of Radiology, Tokai University Hachioji Hospital, 1838 Ishikawa-machi, Hachioji-shi, Tokyo 192-0032, Japan
| | - Ayumi Horikawa
- Department of Transfusion Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Akira Iijima
- Department of Neurosurgery, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Nobuhito Saito
- Department of Neurosurgery, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Koki Takahashi
- Department of Transfusion Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
11
|
Krüger-Genge A, Hiebl B, Franke RP, Lendlein A, Jung F. Effects of Tacrolimus or Sirolimus on the adhesion of vascular wall cells: Controlled in-vitro comparison study. Clin Hemorheol Microcirc 2018; 67:309-318. [PMID: 28869461 DOI: 10.3233/ch-179211] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In drug eluting stents the cytostatic drugs Sirolimus or Tacrolimus are used to inhibit blood vessel restenosis by limiting the proliferation of smooth muscle cells. However, the cytostatic activity of both drugs was shown to be not cell specific and could also affect the stent endothelialisation, respectively. Currently, only limited in vitro data are available about the impact of Sirolimus and Tacrolimus on endothelial cell proliferation over a broad concentration range. To answer this question the following study was performed.Commercially obtained HUVEC were expanded with DMEM cell culture medium (GIBCO, Germany) supplemented with 5 vol% fetal calf serum on non-coated regular polystyrene-based 24-multiwell plates. For drug testings 2×104 cells/cm2 were seeded and grown for 24 h until 30-40% of the multiwell surfaces were covered and then exposed to Sirolimus (1.0×10-11 - 1.0×10-5 mol/l) or Tacrolimus (2.0×10-8 - 6.2×10-5 mol/l), both dissolved in DMSO. 12, 24 and 48 h after adding the drugs cell numbers per area were quantified by counting the cells in six wells with four fields of view per well, representing 0.6 mm2, using a confocal laser microscope.After 48 h of cell growth in the drug-free cell culture medium, the HUVEC number increased from 2.0×104 to 3.55×104 cells/cm2 (mean cell doubling time: 53.6 h, n = 6). At lower concentrations (≤2.0×10-6 mol/l) Tacrolimus reduced the number of adherent HUVEC significantly less than Sirolimus (p < 0.05). However, at higher concentrations (≥2.07×10-5 mol/l) the effect of Tacrolimus on the number of adherent endothelial cells was significantly greater than that of Sirolimus (p < 0.05). At the highest concentration applied (6.22×10-5 mol/l), Tacrolimus induced detachment of all HUVECs within 12 h after drug application. The number of adherent HUVEC decreased only slightly (about 9%) after Sirolimus application at the highest concentration (1.09×10-5 mol/l).These data show that in a non-flow model the cytostatic drug Tacrolimus reduced the number of adherent endothelial cells less than Sirolimus, as long as the drug concentration did not surpass 10-6 mol/l. At the limits of solubility, Sirolimus (1×10-5 mol/l) reduced the number of adherent endothelial cells less than Tacrolimus (6×10-5 mol/l), which induced detachment of endothelial cells.
Collapse
Affiliation(s)
- A Krüger-Genge
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - B Hiebl
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute for Animal Hygiene, Animal Welfare and Farm Animal Behavior, University of Veterinary Medicine Hannover, Hannover, Germany
| | - R P Franke
- University of Ulm, Central Institute for Biomedical Technology, Department of Biomaterials, Ulm, Germany
| | - A Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - F Jung
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| |
Collapse
|
12
|
Autieri MV. IL-19 and Other IL-20 Family Member Cytokines in Vascular Inflammatory Diseases. Front Immunol 2018; 9:700. [PMID: 29681905 PMCID: PMC5897441 DOI: 10.3389/fimmu.2018.00700] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/21/2018] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular disease remains a major medical and socioeconomic burden in developed and developing countries and will increase with an aging and increasingly sedentary society. Many vascular diseases and atherosclerotic vascular disease, in particular, are essentially inflammatory disorders, involving multiple cell types. Communication between these cells is initiated and sustained by a complex network of cytokines and their receptors. The interleukin (IL)-20 family members, IL-19, IL-20, IL-22, and IL-24, initiate, sustain, and drive the progression of vascular disease. They are important in vascular disease as they facilitate a bidirectional cross-talk between resident vascular cells with immune cells. These cytokines are grouped into the same family based on shared common receptor subunits and signaling pathways. This communication is varied and can result in exacerbation, attenuation, and even repair of the vasculature. We will briefly review what is known about IL-20, IL-22, and IL-24 in cardiovascular biology. Because IL-19 is the most studied member of this family in terms of its role in vascular pathophysiological processes, the major emphasis of this review will focus on the expression and atheroprotective roles of IL-19 in vascular inflammatory disease.
Collapse
Affiliation(s)
- Michael V Autieri
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
13
|
Molecular Ultrasound Imaging of αvβ3-Integrin Expression in Carotid Arteries of Pigs After Vessel Injury. Invest Radiol 2017; 51:767-775. [PMID: 27119438 DOI: 10.1097/rli.0000000000000282] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Interventions such as balloon angioplasty can cause vascular injury leading to platelet activation, thrombus formation, and inflammatory response. This induces vascular smooth muscle cell activation and subsequent re-endothelialization with expression of αvβ3-integrin by endothelial cells and vascular smooth muscle cell. Thus, poly-N-butylcyanoacrylate microbubbles (MBs) targeted to αvβ3-integrin were evaluated for monitoring vascular healing after vessel injury in pigs using molecular ultrasound imaging. MATERIALS AND METHODS Approval for animal experiments was obtained. The binding specificity of αvβ3-integrin-targeted MB to human umbilical vein endothelial cells was tested with fluorescence microscopy. In vivo imaging was performed using a clinical ultrasound system and an 8-MHz probe. Six mini pigs were examined after vessel injury in the left carotid artery. The right carotid served as control. Uncoated MB, cDRG-coated MB, and αvβ3-integrin-specific cRGD-coated MB were injected sequentially. Bound MBs were assessed 8 minutes after injection using ultrasound replenishment analysis. Measurements were performed 2 hours, 1 and 5 weeks, and 3 and 6 months after injury. In vivo data were validated by immunohistochemistry. RESULTS Significantly stronger binding of cRGD-MB than MB and cDRG-MB to human umbilical vein endothelial cells was found (P < 0.01). As vessel injury leads to upregulation of αvβ3-integrin, cRGD-MBs bound significantly stronger (P < 0.05) in injured carotid arteries than at the counter side 1 week after vessel injury and significant differences could also be observed after 5 weeks. After 3 months, αvβ3-integrin expression decreased to baseline and binding of cRGD-MB was comparable in both vessels. Values remained at baseline also after 6 months. CONCLUSIONS Ultrasound imaging with RGD-MB is promising for monitoring vascular healing after vessel injury. This may open new perspectives to assess vascular damage after radiological interventions.
Collapse
|
14
|
Han Y, Jiang Q, Wang Y, Li W, Geng M, Han Z, Chen X. The anti-proliferative effects of oleanolic acid on A7r5 cells-Role of UCP2 and downstream FGF-2/p53/TSP-1. Cell Biol Int 2017; 41:1296-1306. [PMID: 28792088 DOI: 10.1002/cbin.10838] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/07/2017] [Indexed: 01/11/2023]
Abstract
Vascular smooth muscle cell (VSMC) proliferation is a major contributor to atherosclerosis. This study investigated the inhibitory effects of oleanolic acid (OA) against oxidized low-density lipoprotein (ox-LDL)-induced VSMC proliferation in A7r5 cells and explored underlying molecular mechanism. The cell proliferation was quantified with cell counting kit-8 (CCK-8), in which ox-LDL significantly increased A7r5 cells proliferation, while OA pretreatment effectively alleviated such changes without inducing overt cytotoxicity, as indicated by lactate dehydrogenase (LDH) assay. Quantitative real-time RT-PCR (qRT-PCR) and Western blotting revealed increased UCP2 and FGF-2 expression levels as well as decreased p53 and TSP-1 expression levels in A7r5 cells following ox-LDL exposure, while OA pretreatment reversed such changes. Furthermore, inhibiting UCP2 with genipin remarkably reversed the changes in the expression levels of FGF-2, p53, and TSP-1 induced by ox-LDL exposure; silencing FGF-2 with siRNA did not significantly change the expression levels of UCP2 but effectively reversed the changes in the expression levels of p53 and TSP-1, and activation of p53 with PRIMA-1 only significantly affected the changes in the expression levels of TSP-1, but not in UCP2 or FGF-2, suggesting a UCP-2/FGF-2/p53/TSP-1 signaling in A7r5 cells response to ox-LDL exposure. Additionally, co-treatment of OA and genipin exhibited similar effects to the expression levels of UCP2, FGF-2, p53, and TSP-1 as OA or genipin solo treatment in ox-LDL-exposed A7r5 cells, suggesting the involvement of UCP-2/FGF-2/p53/TSP-1 in the mechanism of OA. In conclusion, OA inhibits ox-LDL-induced VSMC proliferation in A7r5 cells, the mechanism involves the changes in UCP-2/FGF-2/p53/TSP-1.
Collapse
Affiliation(s)
- Yantao Han
- Qingdao University Medical College, 308 Ningxia Road, Qingdao 266071, Shandong, China
| | - Qixiao Jiang
- Qingdao University Medical College, 308 Ningxia Road, Qingdao 266071, Shandong, China
| | - Yu Wang
- Qingdao University Medical College, 308 Ningxia Road, Qingdao 266071, Shandong, China
| | - Wenqian Li
- Qingdao University Medical College, 308 Ningxia Road, Qingdao 266071, Shandong, China
| | - Min Geng
- Qingdao University Medical College, 308 Ningxia Road, Qingdao 266071, Shandong, China
| | - Zhiwu Han
- The Affiliated Hospital of Qingdao University, 16 Jiansu Road, Qingdao 266021, Shandong, China
| | - Xuehong Chen
- Qingdao University Medical College, 308 Ningxia Road, Qingdao 266071, Shandong, China
| |
Collapse
|
15
|
Abstract
The importance of inflammation and inflammatory pathways in atherosclerotic disease and acute coronary syndromes (ACS) is well established. The success of statin therapy rests not only on potently reducing levels of low-density lipoprotein cholesterol, but also on the many beneficial, pleiotropic effects statin therapy has on various inflammatory mechanisms in atherosclerotic disease, from reducing endothelial dysfunction to attenuating levels of serum C-reactive protein. Due to the growing awareness of the importance of inflammation in ACS, investigators have attempted to develop novel therapies against known markers of inflammation for several decades. Targeted pathways have ranged from inhibiting C5 cleavage with a high-affinity monoclonal antibody against C5 to inhibiting the activation of the p38 mitogen-activated protein kinase signaling cascades. In each of these instances, despite promising early preclinical and mechanistic studies and phase 2 trials suggesting a potential benefit in reducing post-MI complications or restenosis, these novel therapies have failed to show benefits during large, phase 3 clinical outcomes trials. This review discusses several examples of novel anti-inflammatory therapies that failed to show significant improvement on clinical outcomes when tested in large, randomized trials and highlights potential explanations for why targeted therapies against known markers of inflammation in ACS have failed to launch.
Collapse
Key Words
- ACS, acute coronary syndromes
- CABG, coronary artery bypass graft
- CAD, coronary artery disease
- HDL-C, high-density lipoprotein cholesterol
- IL, interleukin
- LDL-C, low-density lipoprotein cholesterol
- Lp-PLA2, lipoprotein-associated phospholipase A2
- MAPK, mitogen-activated protein kinase
- MI, myocardial infarction
- NSTEMI, non–ST-segment myocardial infarction
- PCI, percutaneous coronary intervention
- PSGL, P-selectin glycoprotein ligand
- STEMI, ST-segment elevation myocardial infarction
- SVG, saphenous vein grafts
- TBR, tissue-to-background ratio
- acute coronary syndrome
- anti-inflammatory
- drug targets
- hsCRP, high-sensitivity C-reactive protein
- sPLA2, secretory phospholipase A2
Collapse
|
16
|
Paclitaxel-Coated Balloons: Investigation of Drug Transfer in Healthy and Atherosclerotic Arteries - First Experimental Results in Rabbits at Low Inflation Pressure. Cardiovasc Drugs Ther 2017; 30:263-70. [PMID: 27033233 PMCID: PMC4919377 DOI: 10.1007/s10557-016-6658-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Purpose Beyond antiproliferative properties, paclitaxel exhibits anti-inflammatory activity, which might be beneficial in the local treatment of nonocclusive coronary artery disease. Paclitaxel release and tissue concentrations after paclitaxel-coated balloon treatment using different pressures have not been investigated so far. The aim of the study was to investigate in an atherosclerotic rabbit model whether drug transfer from paclitaxel-coated balloons into the vessel wall is affected by the presence of atherosclerotic lesions and to which extent it depends on the inflation pressure used. Methods Paclitaxel-coated balloons (3.5 μg/mm2 paclitaxel) were inflated with pressures of 1, 2, or 6 atm (60s) in healthy (n = 39) and atherosclerotic (n = 22) arteries of New Zealand White Rabbits. Paclitaxel content in arterial walls (10 min after interventions) and paclitaxel remaining on balloons after treatment were analyzed using high-performance liquid chromatography. Results Median paclitaxel tissue concentrations were 829.3 μg/g (IQR 636.5–1487 μg/g) in healthy and 375.7 μg/g (IQR 169.8–771.6 μg/g) in atherosclerotic arteries (p = 0.0002). The paclitaxel tissue concentration was dependent on inflation pressure (1 atm vs. 2 atm vs. 6 atm) in atherosclerotic arteries (p = 0.0106) but not in healthy arteries (p ≥ 0.05). Conclusions Atherosclerotic lesions impede the transfer of paclitaxel into arterial walls. Higher inflation pressures resulted in an increased paclitaxel transfer in atherosclerotic but not in healthy arteries. However, it is assumed that the tissue concentrations achieved with an inflation pressure of 2 atm are potentially effective in this model.
Collapse
|
17
|
Beltrão-Braga PCB, Koh IHJ, Silva MRR, Gutierrez PS, Han SW. Vascular Adventitia is a Suitable Compartment to Transplant Transduced Vascular Smooth Muscle Cells for Ex Vivo Gene Expression. Cell Transplant 2017. [DOI: 10.3727/000000002783985486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Vascular smooth muscle cells (VSMC) are ideal for systemic gene therapy because of their proximity to blood vessels and they have demonstrated long-term exogenous gene expression in vivo. However, the procedure generally followed to seed the transduced VSMC onto arteries denuded of endothelial cells usually induces stenosis and thrombosis, with a consequent high risk for use in humans. We demonstrate here that the vascular adventitia is a suitable place to introduce transduced VSMC and to secrete therapeutic proteins into the blood stream by a simple procedure, avoiding postoperative vascular complications. Transduced VSMC, with the retroviral vectors carrying the human growth hormone gene (hGH), were seeded into the adventitia of the rat abdominal aorta by single injection of a cell suspension. Based on the hGH and anti-hGH production in serum and on histological analysis of the removed aortas, we demonstrated hGH production over the 2-month experimental period. None of the animals used in the experiment showed stenosis, thrombosis, or other vascular or visible physiological abnormalities.
Collapse
Affiliation(s)
| | - Ivan H. J. Koh
- Department of Pediatrics, UNIFESP-EPM, São Paulo, 04023-062, Brazil
| | | | | | - Sang W. Han
- Department of Biophysics, UNIFESP-EPM, São Paulo, 04023-062, Brazil
| |
Collapse
|
18
|
Nuhn H, Blanco CE, Desai TA. Nanoengineered Stent Surface to Reduce In-Stent Restenosis in Vivo. ACS APPLIED MATERIALS & INTERFACES 2017; 9:19677-19686. [PMID: 28574242 DOI: 10.1021/acsami.7b04626] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In-stent restenosis (ISR) is the leading cause of stent failure and is a direct result of a dysfunctional vascular endothelium and subsequent overgrowth of vascular smooth muscle tissue. TiO2 nanotubular (NT) arrays have been shown to affect vascular endothelial cells (VECs) and vascular smooth muscle cells (VSMCs) in vitro by accelerating VEC cell proliferation and migration while suppressing VSMCs. This study investigates for the first time the potentially beneficial effects of TiO2 NT arrays on vascular tissue in vivo. TiO2 NT arrays (NT diameter: 90 ± 5 nm, height: 1800 ± 300 nm) were grown on the surface of titanium stents and characterized in terms of surface morphology and stability. Stents were implanted into the iliofemoral artery using an overinflation model (rabbit). After 28 days, stenosis rates were determined. The data show a statistically significant reduction of stenosis by 30% compared to the control. Tissue in the presence of TiO2 NTs appears more mature, and less neointima is present between struts. In addition, the extra cellular matrix secreted by cells at the interface of the NT arrays shows complete integration into the nanostructured surface. These results document the accelerated restoration of a functional endothelium in the presence of TiO2 NT arrays and substantiate their beneficial impact on vascular tissue in vivo. Our findings suggest that TiO2 NT arrays can be used as a drug-free approach for keeping stents patent long-term and have the potential to address ISR.
Collapse
Affiliation(s)
- Harald Nuhn
- The Alfred E. Mann Institute for Biomedical Engineering at the University of Southern California , 1042 Downey Way, DRB Building, Suite 101, Los Angeles, California 90089-1112, United States
| | - Cesar E Blanco
- The Alfred E. Mann Institute for Biomedical Engineering at the University of Southern California , 1042 Downey Way, DRB Building, Suite 101, Los Angeles, California 90089-1112, United States
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences and The UC Berkeley-UCSF Graduate Group in Bioengineering, University of California-San Francisco , San Francisco, California 94158, United States
| |
Collapse
|
19
|
Zhang H, Luo H, Sun J, Liu C, Tian Y, Chen H, Zhang C. Mild coronary artery stenosis has no impact on cardiac and vascular parameters in miniature swine exposed to positive acceleration stress. J Cardiovasc Med (Hagerstown) 2017; 17:713-8. [PMID: 25799013 DOI: 10.2459/jcm.0000000000000014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Exposure of pilots' heart to acceleration-associated stress (+Gz stress) is an adverse effect of high-performance aviation. The occurrence of coronary heart diseases is one of the most frequent medical causes leading to cessation of flying. AIM To assess the effects of +Gz stress on coronary artery stenosis (CAS) in a minimally invasive miniature swine model with a fast recovery. METHODS The proximal left anterior descending branch was ligated in 20 swine using silk suture. CAS degree (mild, moderate, severe) was analyzed by quantitative computerized angiography. Five swine underwent a sham operation. +Gz stress exposure was performed and venous blood was collected before/after exposure. Plasma C-reactive protein (CRP), endothelin (ET)-1, angiotensin (Ang) II and urotensin 2 (U2) levels were measured. RESULTS CAS models were successful in 18 animals. Two swine exhibited ventricular fibrillation during the procedure and died. Plasma CRP, ET-1, Ang II and U2 changed significantly after maximal tolerated +Gz stress exposure (all P < 0.05). After maximal tolerated +Gz stress exposure, plasma CRP, ET-1, Ang II and U2 levels increased in the moderate and severe stenosis groups, compared with the sham group (all P < 0.05), but there was no significant difference between the mild stenosis group and the sham group (all P > 0.05). CONCLUSION The fully endoscopic operation method successfully generated animal models of different degrees of CAS. Plasma CRP, ET-1, Ang II and U2 levels increased after +Gz stress exposure with increasing CAS severity. Animals with mild stenosis showed no ill effect under +Gz stress, suggesting that pilots with mild stenosis might be allowed to continue flying, but it must be confirmed in humans.
Collapse
Affiliation(s)
- Haitao Zhang
- aDepartment of Cardiology, Air Force Clinic Institution of Anhui Medical University bDepartment of Cardiology, General Hospital of Air Force, PLA cAnimal Experimental Center of Fuwai Hospital, National Heart Center of China, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
20
|
Lee KP, Kim JE, Kim H, Chang HR, Lee DW, Park WH. Bo-Gan-Whan regulates proliferation and migration of vascular smooth muscle cells. Altern Ther Health Med 2016; 16:306. [PMID: 27549769 PMCID: PMC4994174 DOI: 10.1186/s12906-016-1292-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 08/17/2016] [Indexed: 02/05/2023]
Abstract
Background Bo-Gan-Whan (BGH), a Korean polyherbal medicine, is used as a hepatoprotective drug. It has six natural sources, and has been demonstrated to have anti-oxidative, anti-cancer, and anti-inflammatory properties; however, its effect on vascular diseases remains unclear. Methods Cell viability and proliferation assays were employed using an EZ-Cytox Cell Viability Assay Kit. Platelet-derived growth factor (PDGF)-BB-induced vascular smooth muscle cell (VSMC) migration was measured by scratch wound healing assay and Boyden chamber assay. The expression levels of the phosphorylated signaling proteins relevant to proliferation, including extracellular signal-regulated kinase (ERK) 1/2 and p38 mitogen-activated protein kinase (MAPK) were determined by western blot analysis. Chromatogram and mass analysis were employed by Ultra Performance Liquid Chromatography (UPLC) system. Cell prolife ration and migration were also explored using the PDGF-BB-induced aortic sprout assay. Results BGH (100–500 μg/mL) significantly inhibited the proliferation and migration of PDGF-BB-stimulated VSMCs through the reduced phosphorylation of ERK1/2 and p38 MAPK in comparison to untreated PDGF-BB-stimulated VSMC. Moreover, we identified the paeoniflorin as the major composition of BGH. Conclusions We suggest that BGH may have an anti-atherosclerosis effect by inhibiting the proliferation and migration of PDGF-BB-stimulated VSMCs through down-regulation of ERK1/2 and p38 MAPK phosphorylation.
Collapse
|
21
|
Barone A, Otero-Losada M, Grangeat AM, Cao G, Azzato F, Rodríguez A, Milei J. Ozonetherapy protects from in-stent coronary neointimal proliferation. Role of redoxins. Int J Cardiol 2016; 223:258-261. [PMID: 27541668 DOI: 10.1016/j.ijcard.2016.07.177] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/27/2016] [Indexed: 01/16/2023]
Abstract
BACKGROUND In-stent restenosis and poor re-endothelization usually follow percutaneous transluminal coronary angioplasty, even using drug-eluting stents, due to inflammation and oxidative stress. Medical ozone has antioxidant and anti-inflammatory properties and has not been evaluated in this context. OBJECTIVES To evaluate whether ozonotherapy might reduce restenosis following bare metal stents implantation in relation to the redoxin system in pigs. METHODS Twelve male Landrace pigs (51±9kg) underwent percutaneous transluminal circumflex coronary arteries bare metal stent implantation under heparine infusion and fluoroscopical guidance, using standard techniques. Pigs were randomized to ozonetherapy (n=6) or placebo (n=6) treatment. Before stenting (24h) and twice a week for 30days post-stenting, venous blood was collected, ozonized and reinfused. Same procedure was performed in placebo group except for ozonation. Both groups received antiplatelet treatment. Histopathology and immunohistochemistry studies were performed. RESULTS Severe inflammatory reaction and restenosis with increase in the immunohistochemical expression of thioredoxin-1 were observed in placebo group 30days after surgery. Oppositely, ozonetherapy drastically reduced inflammatory reaction and restenosis, and showed no increase in the Trx-1 immunohistochemical expression 30days after surgery. Immunolabeling for Prx-2 was negative in both groups. Ozonated autohemotherapy strikingly reduced restenosis 30days following PTCA with BMS implantation in pigs. CONCLUSIONS Stimulation of the redoxin system by ozone pretreatment might neutralize oxidative damage from the start and increase antioxidative buffering capacity post-injury, reducing further damage and so the demand for antioxidant enzymes. Our interpretation agrees with the ozone oxidative preconditioning mechanism, extensively investigated.
Collapse
Affiliation(s)
- A Barone
- Institute of Cardiological Research, University of Buenos Aires, National Research Council Argentina, ININCA-UBA-CONICET, Argentina
| | - M Otero-Losada
- Institute of Cardiological Research, University of Buenos Aires, National Research Council Argentina, ININCA-UBA-CONICET, Argentina..
| | - A M Grangeat
- Institute of Cardiological Research, University of Buenos Aires, National Research Council Argentina, ININCA-UBA-CONICET, Argentina
| | - G Cao
- Institute of Cardiological Research, University of Buenos Aires, National Research Council Argentina, ININCA-UBA-CONICET, Argentina
| | - F Azzato
- Institute of Cardiological Research, University of Buenos Aires, National Research Council Argentina, ININCA-UBA-CONICET, Argentina
| | - A Rodríguez
- Otamendi Hospital, Post Graduate School of Medicine, Cardiac Unit, Buenos Aires, Argentina
| | - J Milei
- Institute of Cardiological Research, University of Buenos Aires, National Research Council Argentina, ININCA-UBA-CONICET, Argentina
| |
Collapse
|
22
|
Fontaine AB, Passos SD, Spigos D, Cearlock J, Urbaneja A. Use of Polyetherurethane to Improve the Biocompatibility of Vascular Stents. J Endovasc Ther 2016. [DOI: 10.1177/152660289500200304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Purpose: To investigate potential differential growth of neointima following overdilating arterial trauma with polyetherurethane-coated versus bare metalìc stents in swine. Methods: Twelve specially constructed tantalum stents, 6 coated with polyetherurethane block copolymer and 6 uncoated, were overdilated by 25% in 12 normal renal arteries of six swine. The stents were harvested 8 weeks after implantation and prepared for histologic examination. Neointimal thickness was quantified and analyzed for significant differences between coated and uncoated prostheses. Results: All specimens demonstrated fractures of the internal elastic lamina consistent with vascular injury. There was significantly less neointimal formation (0.0001 < p < 0.05) in coated specimens as compared to uncoated controls in each test animal. Conclusion: The vascular response to overdilating stent trauma appears to be moderated with the use of polyetherurethane block copolymer as compared to control.
Collapse
Affiliation(s)
| | | | | | - Jody Cearlock
- Department of Radiology, Ohio State University Hospitals
| | | |
Collapse
|
23
|
Gupta GK, Agrawal T, Rai V, Del Core MG, Hunter WJ, Agrawal DK. Vitamin D Supplementation Reduces Intimal Hyperplasia and Restenosis following Coronary Intervention in Atherosclerotic Swine. PLoS One 2016; 11:e0156857. [PMID: 27271180 PMCID: PMC4894559 DOI: 10.1371/journal.pone.0156857] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/21/2016] [Indexed: 11/18/2022] Open
Abstract
Vitamin D is a fat-soluble steroid hormone that activates vitamin D receptor to regulate multiple downstream signaling pathways and transcription of various target genes. There is an association between vitamin D deficiency and increased risk for cardiovascular disease. However, most of the studies are observational and associative in nature with limited data on clinical application. Thus, there is a need for more prospective randomized controlled studies to determine whether or not vitamin D supplementation provides cardiovascular protection. In this study, we examined the effects of the deficiency and supplementation of vitamin D on coronary restenosis following coronary intervention in atherosclerotic Yucatan microswine. Twelve Yucatan microswine were fed vitamin D-deficient (n = 4) or -sufficient (n = 8) high cholesterol diet for 6-months followed by coronary intervention. Post-intervention, swine in the vitamin D-sufficient high cholesterol diet group received daily oral supplementation of either 1,000 IU (n = 4) or 3,000 IU (n = 4) vitamin D3. Six months later, optical coherence tomography (OCT) was performed to monitor the development of intimal hyperplasia and restenosis. Animals were euthanized to isolate arteries for histomorphometric and immunohistochemical studies. Animals had graded levels of serum 25(OH)D; vitamin D-deficient (15.33 ± 1.45 ng/ml), vitamin D-sufficient + 1,000 IU oral vitamin D post-intervention (32.27 ± 1.20 ng/ml), and vitamin D-sufficient + 3,000 IU oral vitamin D post-intervention (51.00 ± 3.47 ng/ml). Findings from the OCT and histomorphometric studies showed a decrease in intimal hyperplasia and restenosis in vitamin D-supplemented compared to vitamin D-deficient swine. Vitamin D supplementation significantly decreased serum levels of TNF-α and IFN-γ, upregulated serum levels of IL-10, and had no effect on serum IL-6 levels. These findings suggest that vitamin D supplementation limits neointimal formation following coronary intervention in atherosclerotic swine and provide the support for vitamin D supplementation to protect against the development of coronary restenosis.
Collapse
Affiliation(s)
- Gaurav K. Gupta
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States of America
| | - Tanupriya Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States of America
| | - Vikrant Rai
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States of America
| | - Michael G. Del Core
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States of America
| | - William J. Hunter
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States of America
| | - Devendra K. Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States of America
- * E-mail:
| |
Collapse
|
24
|
Rusin CG, Acosta SI, Shekerdemian LS, Vu EL, Bavare AC, Myers RB, Patterson LW, Brady KM, Penny DJ. Prediction of imminent, severe deterioration of children with parallel circulations using real-time processing of physiologic data. J Thorac Cardiovasc Surg 2016; 152:171-7. [PMID: 27174513 DOI: 10.1016/j.jtcvs.2016.03.083] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 03/02/2016] [Accepted: 03/20/2016] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Sudden death is common in patients with hypoplastic left heart syndrome and comparable lesions with parallel systemic and pulmonary circulation from a common ventricular chamber. It is hypothesized that unforeseen acute deterioration is preceded by subtle changes in physiologic dynamics before overt clinical extremis. Our objective was to develop a computer algorithm to automatically recognize precursors to deterioration in real-time, providing an early warning to care staff. METHODS Continuous high-resolution physiologic recordings were obtained from 25 children with parallel systemic and pulmonary circulation who were admitted to the cardiovascular intensive care unit of Texas Children's Hospital between their early neonatal palliation and stage 2 surgical palliation. Instances of cardiorespiratory deterioration (defined as the need for cardiopulmonary resuscitation or endotracheal intubation) were found via a chart review. A classification algorithm was applied to both primary and derived parameters that were significantly associated with deterioration. The algorithm was optimized to discriminate predeterioration physiology from stable physiology. RESULTS Twenty cardiorespiratory deterioration events were identified in 13 of the 25 infants. The resulting algorithm was both sensitive and specific for detecting impending events, 1 to 2 hours in advance of overt extremis (receiver operating characteristic area = 0.91, 95% confidence interval = 0.88-0.94). CONCLUSIONS Automated, intelligent analysis of standard physiologic data in real-time can detect signs of clinical deterioration too subtle for the clinician to observe without the aid of a computer. This metric may serve as an early warning indicator of critical deterioration in patients with parallel systemic and pulmonary circulation.
Collapse
Affiliation(s)
- Craig G Rusin
- Department of Pediatrics-Cardiology, Baylor College of Medicine, Texas Children's Hospital, Houston, Tex.
| | - Sebastian I Acosta
- Department of Pediatrics-Cardiology, Baylor College of Medicine, Texas Children's Hospital, Houston, Tex
| | - Lara S Shekerdemian
- Department of Pediatrics-Critical Care, Baylor College of Medicine, Texas Children's Hospital, Houston, Tex
| | - Eric L Vu
- Department of Pediatrics-Anesthesia, Baylor College of Medicine, Texas Children's Hospital, Houston, Tex
| | - Aarti C Bavare
- Department of Pediatrics-Critical Care, Baylor College of Medicine, Texas Children's Hospital, Houston, Tex
| | - Risa B Myers
- Department of Computer Science, Rice University, Houston, Tex
| | - Lance W Patterson
- Department of Pediatrics-Cardiology, Baylor College of Medicine, Texas Children's Hospital, Houston, Tex
| | - Ken M Brady
- Department of Pediatrics-Anesthesia, Baylor College of Medicine, Texas Children's Hospital, Houston, Tex
| | - Daniel J Penny
- Department of Pediatrics-Cardiology, Baylor College of Medicine, Texas Children's Hospital, Houston, Tex
| |
Collapse
|
25
|
Tsang HG, Rashdan NA, Whitelaw CBA, Corcoran BM, Summers KM, MacRae VE. Large animal models of cardiovascular disease. Cell Biochem Funct 2016; 34:113-32. [PMID: 26914991 PMCID: PMC4834612 DOI: 10.1002/cbf.3173] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 01/26/2016] [Accepted: 01/27/2016] [Indexed: 12/12/2022]
Abstract
The human cardiovascular system is a complex arrangement of specialized structures with distinct functions. The molecular landscape, including the genome, transcriptome and proteome, is pivotal to the biological complexity of both normal and abnormal mammalian processes. Despite our advancing knowledge and understanding of cardiovascular disease (CVD) through the principal use of rodent models, this continues to be an increasing issue in today's world. For instance, as the ageing population increases, so does the incidence of heart valve dysfunction. This may be because of changes in molecular composition and structure of the extracellular matrix, or from the pathological process of vascular calcification in which bone-formation related factors cause ectopic mineralization. However, significant differences between mice and men exist in terms of cardiovascular anatomy, physiology and pathology. In contrast, large animal models can show considerably greater similarity to humans. Furthermore, precise and efficient genome editing techniques enable the generation of tailored models for translational research. These novel systems provide a huge potential for large animal models to investigate the regulatory factors and molecular pathways that contribute to CVD in vivo. In turn, this will help bridge the gap between basic science and clinical applications by facilitating the refinement of therapies for cardiovascular disease.
Collapse
Affiliation(s)
- H G Tsang
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, SCT, UK
| | - N A Rashdan
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, SCT, UK
| | - C B A Whitelaw
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, SCT, UK
| | - B M Corcoran
- Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, SCT, UK
| | - K M Summers
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, SCT, UK
| | - V E MacRae
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, SCT, UK
| |
Collapse
|
26
|
Lee HF, Wu LS, Chan YH, Lee CH, Liu JR, Tu HT, Wen MS, Kuo CT, Chen WJ, Yeh YH, See LC, Chang SH. Dialysis Patients with Implanted Drug-Eluting Stents Have Lower Major Cardiac Events and Mortality than Those with Implanted Bare-Metal Stents: A Taiwanese Nationwide Cohort Study. PLoS One 2016; 11:e0146343. [PMID: 26731408 PMCID: PMC4711720 DOI: 10.1371/journal.pone.0146343] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/15/2015] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE To investigate the efficacy and long-term clinical benefits of DES for dialysis patients. BACKGROUND It is unclear whether percutaneous coronary intervention (PCI) with drug-eluting stent (DES) implantation is associated with lower rates of major adverse cardiovascular events (MACE) or mortality compared to bare-metal stents (BMS). METHODS From a nationwide cohort selected from Taiwan's National Health Insurance Research Database, we enrolled 2,835 dialysis patients who were hospitalized for PCI treatment with stent implantation from Dec 1, 2006. Follow-up was from the date of index hospitalization for PCI until the first MACE, date of death, or December 31, 2011, whichever came first. RESULTS A total of 738 patients (26.0%) had DES implanted, and 2,097 (74%) had BMS implanted. The medium time to the first MACE was 0.53 years (interquartile range: 0.89 years; range: 0-4.62 years). At 1-year follow-up, patients treated with BMS had significantly, non-fatal myocardial infarction (MI), all-cause mortality, and composite MACE compared to those treated with DES. The overall repeat revascularization with coronary artery bypass graft (CABG), non-fatal MI, all-cause mortality, and composite MACE were significantly lower in patients treated with DES than those treated with BMS. Multivariate cox regression analysis showed that older age, history of diabetes, history of heart failure, history of stroke, and DES vs. BMS were independent significant predictors of MACE. CONCLUSIONS DES implantation conferred survival benefits in dialysis patients compared with BMS implantation.
Collapse
Affiliation(s)
- Hsin-Fu Lee
- Chang Gung University and Department of Cardiology, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Lung-Sheng Wu
- Chang Gung University and Department of Cardiology, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Yi-Hsin Chan
- Chang Gung University and Department of Cardiology, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Cheng-Hung Lee
- Chang Gung University and Department of Cardiology, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Jia-Rou Liu
- Department of Public Health, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hui-Tzu Tu
- Department of Public Health, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ming-Shien Wen
- Chang Gung University and Department of Cardiology, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Chi-Tai Kuo
- Chang Gung University and Department of Cardiology, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Wei-Jan Chen
- Chang Gung University and Department of Cardiology, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Yung-Hsin Yeh
- Chang Gung University and Department of Cardiology, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Lai-Chu See
- Department of Public Health, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Biostatistics Core Laboratory, Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
- * E-mail: (LCS); (SHC)
| | - Shang-Hung Chang
- Chang Gung University and Department of Cardiology, Chang Gung Memorial Hospital, Taipei, Taiwan
- * E-mail: (LCS); (SHC)
| |
Collapse
|
27
|
Swier VJ, Tang L, Krueger KD, Radwan MM, Del Core MG, Agrawal DK. Coronary Injury Score Correlates with Proliferating Cells and Alpha-Smooth Muscle Actin Expression in Stented Porcine Coronary Arteries. PLoS One 2015; 10:e0138539. [PMID: 26382957 PMCID: PMC4575201 DOI: 10.1371/journal.pone.0138539] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 08/31/2015] [Indexed: 11/30/2022] Open
Abstract
Neointimal formation and cell proliferation resulting into in-stent restenosis is a major pathophysiological event following the deployment of stents in the coronary arteries. In this study, we assessed the degree of injury, based on damage to internal elastic lamina, media, external elastic lamina, and adventitia following the intravascular stenting, and its relationship with the degree of smooth muscle cell proliferation. We examined the smooth muscle cell proliferation and their phenotype at different levels of stent injury in the coronary arteries of domestic swine fed a normal swine diet. Five weeks after stent implantation, swine with and without stents were euthanized and coronaries were excised. Arteries were embedded in methyl methacrylate and sections were stained with H&E, trichrome, and Movat’s pentachrome. The expression of Ki67, α-smooth muscle actin (SMA), vimentin, and HMGB1 was evaluated by immunofluorescence. There was a positive correlation between percent area stenosis and injury score. The distribution of SMA and vimentin was correlated with the degree of arterial injury such that arteries that had an injury score >2 did not have immunoreactivity to SMA in the neointimal cells near the stent struts, but these neointimal cells were positive for vimentin, suggesting a change in the smooth muscle cell phenotype. The Ki67 and HMGB1 immunoreactivity was highly correlated with the fragmentation of the IEL and injury in the tunica media. Thus, the extent of coronary arterial injury during interventional procedure will dictate the degree of neointimal hyperplasia, in-stent restenosis, and smooth muscle cell phenotype.
Collapse
Affiliation(s)
- Vicki J. Swier
- Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, United States of America
| | - Lin Tang
- Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, United States of America
| | - Kristopher D. Krueger
- Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, United States of America
| | - Mohamed M. Radwan
- Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, United States of America
| | - Michael G. Del Core
- Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, United States of America
| | - Devendra K. Agrawal
- Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
28
|
Lim S, Lee KS, Lee JE, Park HS, Kim KM, Moon JH, Choi SH, Park KS, Kim YB, Jang HC. Effect of a new PPAR-gamma agonist, lobeglitazone, on neointimal formation after balloon injury in rats and the development of atherosclerosis. Atherosclerosis 2015; 243:107-19. [PMID: 26363808 DOI: 10.1016/j.atherosclerosis.2015.08.037] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 08/17/2015] [Accepted: 08/26/2015] [Indexed: 12/25/2022]
Abstract
OBJECTIVE The ligand-activated transcription factor peroxisome proliferator-activated receptor gamma (PPARγ) is a key factor in adipogenesis, insulin sensitivity, and cell cycle regulation. Activated PPARγ might also have anti-inflammatory and antiatherogenic properties. We tested whether lobeglitazone, a new PPARγ agonist, might protect against atherosclerosis. METHODS A rat model of balloon injury to the carotid artery, and high-fat, high-cholesterol diet-fed apolipoprotein E gene knockout (ApoE(-/-)) mice were studied. RESULTS After the balloon injury, lobeglitazone treatment (0.3 and 0.9 mg/kg) caused a significant decrease in the intima-media ratio compared with control rats (2.2 ± 0.9, 1.8 ± 0.8, vs. 3.3 ± 1.2, P < 0.01). Consistent with this, in ApoE(-/-) mice fed a high-fat diet, lobeglitazone treatment (1, 3, and 10 mg/kg) for 8 weeks reduced atherosclerotic lesion sizes in the aorta compared with the control mice in a dose-dependent manner. Treatment of vascular smooth muscle cells with lobeglitazone inhibited proliferation and migration and blocked the cell cycle G0/G1 to S phase progression dose-dependently. In response to lobeglitazone, tumor necrosis factor alpha (TNFα)-induced monocyte-endothelial cell adhesion was decreased by downregulating the levels of adhesion molecules. TNFα-induced nuclear factor kappa-B (NF-κB) p65 translocation into the nucleus was also blocked in endothelial cells. Insulin resistance was decreased by lobeglitazone treatment. Circulating levels of high sensitivity C-reactive protein and monocyte chemoattractant protein-1 were decreased while adiponectin levels were increased by lobeglitazone in the high-fat diet-fed ApoE(-/-) mice. CONCLUSION Lobeglitazone has antiatherosclerotic properties and has potential for treating patients with diabetes and cardiovascular risk.
Collapse
Affiliation(s)
- Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-070, South Korea; Department of Internal Medicine, Seoul National University College of Medicine, 28 Yongon-Dong, Chongno-Gu, Seoul, 110-744, South Korea
| | - Kuy-Sook Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-070, South Korea; Biomedical Research Institute, Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-070, South Korea
| | - Jie Eun Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-070, South Korea; Department of Internal Medicine, Seoul National University College of Medicine, 28 Yongon-Dong, Chongno-Gu, Seoul, 110-744, South Korea
| | - Ho Seon Park
- Department of Internal Medicine, Seoul National University College of Medicine, 28 Yongon-Dong, Chongno-Gu, Seoul, 110-744, South Korea
| | - Kyoung Min Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-070, South Korea; Department of Internal Medicine, Seoul National University College of Medicine, 28 Yongon-Dong, Chongno-Gu, Seoul, 110-744, South Korea
| | - Jae Hoon Moon
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-070, South Korea; Department of Internal Medicine, Seoul National University College of Medicine, 28 Yongon-Dong, Chongno-Gu, Seoul, 110-744, South Korea
| | - Sung Hee Choi
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-070, South Korea; Department of Internal Medicine, Seoul National University College of Medicine, 28 Yongon-Dong, Chongno-Gu, Seoul, 110-744, South Korea
| | - Kyong Soo Park
- Department of Internal Medicine, Seoul National University College of Medicine, 28 Yongon-Dong, Chongno-Gu, Seoul, 110-744, South Korea; Biomedical Research Institute, Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-070, South Korea
| | - Young Bum Kim
- Biomedical Research Institute, Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-070, South Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 110-744, South Korea; Division of Endocrinology, Metabolism and Diabetes, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Boston, MA, 02215, USA
| | - Hak Chul Jang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-070, South Korea; Department of Internal Medicine, Seoul National University College of Medicine, 28 Yongon-Dong, Chongno-Gu, Seoul, 110-744, South Korea.
| |
Collapse
|
29
|
Yao W, Sun Q, Huang L, Meng G, Wang H, Jing X, Zhang W. Tetrahydroxystilbene glucoside inhibits TNF-α-induced migration of vascular smooth muscle cells via suppression of vimentin. Can J Physiol Pharmacol 2015; 94:155-160. [PMID: 26583578 DOI: 10.1139/cjpp-2015-0160] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Vascular smooth muscle cell (VSMC) migration triggered by TNF-α is an important event that occurs during the development of atherosclerosis. 2,3,5,4'-Tetrahydroxystilbene-2-O-β-d-glucoside (TSG) has been proven to exhibit significant anti-atherosclerotic activity. Herein we investigate the inhibitory effect of TSG on TNF-α-induced VSMC migration and explore the underlying mechanisms. TSG pretreatment markedly inhibited TNF-α-induced cell migration. The inhibition of vimentin redistribution and expression was involved in the inhibitory effect of TSG on VSMC migration. The suppression of vimentin expression by shRNA in VSMCs significantly inhibited TNF-α-induced cell migration. Furthermore, TSG inhibited the TNF-α-induced expression of TGFβ1 and TGFβR1, and phosphorylation of TGFβR1 and Smad2/3. TSG also suppressed the nuclear translocation of Smad4 induced by TNF-α. These results suggest that TSG inhibits VSMC migration induced by TNF-α through inhibiting vimentin rearrangement and expression. The interruption of TGFβ/Smad pathway appears to be responsible for the suppression of TSG on vimentin expression.
Collapse
Affiliation(s)
- Wenjuan Yao
- Department of Pharmacology, School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, P. R. China.,Department of Pharmacology, School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, P. R. China
| | - Qinju Sun
- Department of Pharmacology, School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, P. R. China.,Department of Pharmacology, School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, P. R. China
| | - Lei Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, P. R. China.,Department of Pharmacology, School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, P. R. China
| | - Guoliang Meng
- Department of Pharmacology, School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, P. R. China.,Department of Pharmacology, School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, P. R. China
| | - Huiming Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, P. R. China.,Department of Pharmacology, School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, P. R. China
| | - Xiang Jing
- Department of Pharmacology, School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, P. R. China.,Department of Pharmacology, School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, P. R. China
| | - Wei Zhang
- Department of Pharmacology, School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, P. R. China.,Department of Pharmacology, School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, P. R. China
| |
Collapse
|
30
|
Chen YC, Wen ZH, Lee YH, Chen CL, Hung HC, Chen CH, Chen WF, Tsai MC. Dihydroaustrasulfone alcohol inhibits PDGF-induced proliferation and migration of human aortic smooth muscle cells through inhibition of the cell cycle. Mar Drugs 2015; 13:2390-406. [PMID: 25898413 PMCID: PMC4413217 DOI: 10.3390/md13042390] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 03/27/2015] [Accepted: 04/09/2015] [Indexed: 12/26/2022] Open
Abstract
Dihydroaustrasulfone alcohol is the synthetic precursor of austrasulfone, which is a marine natural product, isolated from the Taiwanese soft coral Cladiella australis. Dihydroaustrasulfone alcohol has anti-inflammatory, neuroprotective, antitumor and anti-atherogenic properties. Although dihydroaustrasulfone alcohol has been shown to inhibit neointima formation, its effect on human vascular smooth muscle cells (VSMCs) has not been elucidated. We examined the effects and the mechanisms of action of dihydroaustrasulfone alcohol on proliferation, migration and phenotypic modulation of human aortic smooth muscle cells (HASMCs). Dihydroaustrasulfone alcohol significantly inhibited proliferation, DNA synthesis and migration of HASMCs, without inducing cell death. Dihydroaustrasulfone alcohol also inhibited platelet-derived growth factor (PDGF)-induced expression of cyclin-dependent kinases (CDK) 2, CDK4, cyclin D1 and cyclin E. In addition, dihydroaustrasulfone alcohol inhibited PDGF-induced phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), whereas it had no effect on the phosphorylation of phosphatidylinositol 3-kinase (PI3K)/(Akt). Moreover, treatment with PD98059, a highly selective ERK inhibitor, blocked PDGF-induced upregulation of cyclin D1 and cyclin E and downregulation of p27kip1. Furthermore, dihydroaustrasulfone alcohol also inhibits VSMC synthetic phenotype formation induced by PDGF. For in vivo studies, dihydroaustrasulfone alcohol decreased smooth muscle cell proliferation in a rat model of restenosis induced by balloon injury. Immunohistochemical staining showed that dihydroaustrasulfone alcohol noticeably decreased the expression of proliferating cell nuclear antigen (PCNA) and altered VSMC phenotype from a synthetic to contractile state. Our findings provide important insights into the mechanisms underlying the vasoprotective actions of dihydroaustrasulfone alcohol and suggest that it may be a useful therapeutic agent for the treatment of vascular occlusive disease.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/administration & dosage
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Aorta/cytology
- Butanones/administration & dosage
- Butanones/pharmacology
- Butanones/therapeutic use
- Cardiovascular Agents/administration & dosage
- Cardiovascular Agents/pharmacology
- Cardiovascular Agents/therapeutic use
- Carotid Artery Injuries/drug therapy
- Carotid Artery Injuries/immunology
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/pathology
- Carotid Artery, Common/drug effects
- Carotid Artery, Common/immunology
- Carotid Artery, Common/metabolism
- Carotid Artery, Common/pathology
- Cell Cycle/drug effects
- Cell Cycle Proteins/antagonists & inhibitors
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Female
- Gene Expression Regulation/drug effects
- Humans
- Injections, Intraperitoneal
- MAP Kinase Signaling System/drug effects
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/immunology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Platelet-Derived Growth Factor/antagonists & inhibitors
- Platelet-Derived Growth Factor/metabolism
- Rats, Sprague-Dawley
- Sulfones/administration & dosage
- Sulfones/pharmacology
- Sulfones/therapeutic use
Collapse
Affiliation(s)
- Yao-Chang Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Lienhai Road, Kaohsiung 804, Taiwan.
- Department of Biomedical Engineering, National Defense Medical Center, Sec. 6, Minquan E. Road, Taipei 11490, Taiwan.
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Lienhai Road, Kaohsiung 804, Taiwan.
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung 80424, Taiwan.
| | - Yen-Hsien Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei 11042, Taiwan.
| | - Chu-Lun Chen
- Department of Physiology and Biophysics; Graduate Institute of Physiology, National Defense Medical Center, Sec. 6, Minquan E. Road, Taipei 11490, Taiwan.
| | - Han-Chun Hung
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung 80424, Taiwan.
| | - Chun-Hong Chen
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung 80424, Taiwan.
| | - Wu-Fu Chen
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Min-Chien Tsai
- Department of Physiology and Biophysics; Graduate Institute of Physiology, National Defense Medical Center, Sec. 6, Minquan E. Road, Taipei 11490, Taiwan.
| |
Collapse
|
31
|
Goodfriend AC, Welch TR, Barker G, Ginther R, Riegel MS, Reddy SV, Wang J, Nugent A, Forbess J. Novel bioresorbable stent coating for drug release in congenital heart disease applications. J Biomed Mater Res A 2014; 103:1761-70. [PMID: 25196819 DOI: 10.1002/jbm.a.35313] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 05/06/2014] [Accepted: 08/14/2014] [Indexed: 11/09/2022]
Abstract
A novel double opposed helical poly-l-lactic acid (PLLA) bioresorbable stent has been designed for use in pediatrics. The aim was to test the PLLA stent biocompatibility. The PLLA stent was immersed into whole pig's blood in a closed loop circuit then fibrin and platelet association was assessed via enzyme-linked immunosorbent assay. D-Dimer was valued at 0.2 ± 0.002 ng/mL and P-selectin 0.43 ± 00.01 ng/mL indicating limited association of fibrin and platelets on the stent. To improve biocompatibility by targeting inflammatory cells, dexamethasone was incorporated on PLLA fibers with two coating methods. Both coatings were poly(l-lactide-co-glycolide) acid (PLGA) but one was made porous with sucrose while the other remained nonporous. There was no change in mechanical properties of the fiber with either coating of PLGA polymer. The total amount of dexamethasone released was then determined for each coating. The cumulative drug release for the porous fiber was significantly higher (∼100%) over 8 weeks than the nonporous fiber (40%). Surface examination of the fiber with scanning electron microscopy showed more surface microfracturing in coatings that contain pores. The biocompatibility of this novel stent was demonstrated. Mechanical properties of the fiber were not altered by coating with PLGA polymer. Anti-inflammatory drug release was optimized using a porous PLGA polymer.
Collapse
Affiliation(s)
- Amy C Goodfriend
- Department of Pediatric Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, 75390
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Ellison S, Gabunia K, Richards JM, Kelemen SE, England RN, Rudic D, Azuma YT, Munroy MA, Eguchi S, Autieri MV. IL-19 reduces ligation-mediated neointimal hyperplasia by reducing vascular smooth muscle cell activation. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2134-43. [PMID: 24814101 DOI: 10.1016/j.ajpath.2014.04.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/31/2014] [Accepted: 04/07/2014] [Indexed: 11/29/2022]
Abstract
We tested the hypothesis that IL-19, a putative member of the type 2 helper T-cell family of anti-inflammatory interleukins, can attenuate intimal hyperplasia and modulate the vascular smooth muscle cell (VSMC) response to injury. Ligated carotid artery of IL-19 knockout (KO) mice demonstrated a significantly higher neointima/intima ratio compared with wild-type (WT) mice (P = 0.04). More important, the increased neointima/intima ratio in the KO could be reversed by injection of 10 ng/g per day recombinant IL-19 into the KO mouse (P = 0.04). VSMCs explanted from IL-19 KO mice proliferated significantly more rapidly than WT. This could be inhibited by addition of IL-19 to KO VSMCs (P = 0.04 and P < 0.01). IL-19 KO VSMCs migrated more rapidly compared with WT (P < 0.01). Interestingly, there was no type 1 helper T-cell polarization in the KO mouse, but there was significantly greater leukocyte infiltrate in the ligated artery in these mice compared with WT. IL-19 KO VSMCs expressed significantly greater levels of inflammatory mRNA, including IL-1β, tumor necrosis factor α, and monocyte chemoattractant protein-1 in response to tumor necrosis factor α stimulation (P < 0.01 for all). KO VSMCs expressed greater adhesion molecule expression and adherence to monocytes. Together, these data indicate that IL-19 is a previously unrecognized counterregulatory factor for VSMCs, and its expression is an important protective mechanism in regulation of vascular restenosis.
Collapse
Affiliation(s)
- Stephen Ellison
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Khatuna Gabunia
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - James M Richards
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Sheri E Kelemen
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Ross N England
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Dan Rudic
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, Georgia
| | - Yasu-Taka Azuma
- Laboratory of Veterinary Pharmacology, Osaka Prefecture University Graduate School, Osaka, Japan
| | - M Alexandra Munroy
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Satoru Eguchi
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Michael V Autieri
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
33
|
Ding Y, Yang KD, Yang Q. The role of PPARδ signaling in the cardiovascular system. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 121:451-73. [PMID: 24373246 DOI: 10.1016/b978-0-12-800101-1.00014-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARα, β/δ, and γ), members of the nuclear receptor transcription factor superfamily, play important roles in the regulation of metabolism, inflammation, and cell differentiation. All three PPAR subtypes are expressed in the cardiovascular system with various expression patterns. Among the three PPAR subtypes, PPARδ is the least studied but has arisen as a potential therapeutic target for cardiovascular and many other diseases. It is known that PPARδ is ubiquitously expressed and abundantly expressed in cardiomyocytes. Accumulated evidence illustrates the role of PPARδ in regulating cardiovascular function and determining pathological progression. In this chapter, we will discuss the current knowledge in the role of PPARδ in the cardiovascular system, the mechanistic insights, and the potential therapeutic utilization for treating cardiovascular disease.
Collapse
Affiliation(s)
- Yishu Ding
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kevin D Yang
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Qinglin Yang
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
34
|
Tse BC, Steinle JJ, Johnson D, Haik BG, Wilson MW. Superselective intraophthalmic artery chemotherapy in a nonhuman primate model: histopathologic findings. JAMA Ophthalmol 2013; 131:903-11. [PMID: 23619956 DOI: 10.1001/jamaophthalmol.2013.2065] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
IMPORTANCE We describe the histopathologic findings in a nonhuman primate (NHP) model of superselective intraophthalmic artery chemotherapy (SSIOAC), detailing ocular and orbital vascular adverse effects. OBJECTIVE To further document, using comprehensive ocular and orbital histopathology, previously reported toxic effects observed with real-time ophthalmoscopy during SSIOAC in a NHP model. DESIGN Comparative interventional case series. SETTING Preclinical trial approved under the guidelines of the Institutional Animal Care and Utilization committee. PARTICIPANTS Six adult male rhesus macaques (Macacca mulatta). INTERVENTIONS The right eye of each NHP was treated with 3 cycles of SSIOAC using either melphalan (5 mg/30 mL) or carboplatin (30 mg/30 mL). Both eyes in each animal were enucleated 6 hours after the final procedure, before euthanasia and formalin perfusion of the NHP; we then performed orbital dissection of the arterial vasculature and optic nerves. MAIN OUTCOME MEASURES Histopathologic examination of the eyes, optic nerves, and orbital vessels of the 6 treated NHPs. RESULTS We found leukostasis with retinal arteriole occlusion in all treated eyes. Retinal endothelial cells stained positive for 2 inflammatory markers, intercellular adhesion molecule 1 and interleukin 8. Transmission electron microscopy revealed occlusion of the retinal vessels with ultrastructural changes in the endothelial cells and surrounding pericytes. Additional findings included nerve fiber layer infarcts, central retinal artery thrombosis, hypertrophy and occlusion of choroidal arteries with disruption of the internal elastic lamina, patchy choroidal inflammation, and birefringent intravascular foreign bodies. Orbital findings included ophthalmic artery and central retinal artery wall dissection, fracturing of the internal elastic lamina, intimal hyperplasia, and eyelid vessel damage. Optic nerves displayed hemorrhage, leukostasis, and foreign body crystallization. Control eyes, optic nerves, and orbital vessels were normal. CONCLUSIONS AND RELEVANCE Histopathologic examination of our nonhuman primate model for SSIOAC revealed significant toxic effects in the ocular and orbital vasculature. These findings substantiate previous observations with real-time retinal imaging and parallel reported vascular toxic effects in children with retinoblastoma treated with SSIOAC.
Collapse
Affiliation(s)
- Brian C Tse
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | | | | | | | | |
Collapse
|
35
|
Autieri MV. Increasing our IQ of vascular smooth muscle cell migration with IQGAP1. Focus on "IQGAP1 links PDGF receptor-β signal to focal adhesions involved in vascular smooth muscle cell migration: role in neointimal formation after vascular injury". Am J Physiol Cell Physiol 2013; 305:C579-80. [PMID: 23657571 DOI: 10.1152/ajpcell.00125.2013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Michael V Autieri
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
36
|
Compound K, an intestinal metabolite of ginsenosides, inhibits PDGF-BB-induced VSMC proliferation and migration through G1 arrest and attenuates neointimal hyperplasia after arterial injury. Atherosclerosis 2013; 228:53-60. [DOI: 10.1016/j.atherosclerosis.2013.02.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 01/29/2013] [Accepted: 02/03/2013] [Indexed: 11/22/2022]
|
37
|
Flege C, Vogt F, Höges S, Jauer L, Borinski M, Schulte VA, Hoffmann R, Poprawe R, Meiners W, Jobmann M, Wissenbach K, Blindt R. Development and characterization of a coronary polylactic acid stent prototype generated by selective laser melting. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2013; 24:241-255. [PMID: 23053808 DOI: 10.1007/s10856-012-4779-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Accepted: 09/25/2012] [Indexed: 06/01/2023]
Abstract
In-stent restenosis is still an important issue and stent thrombosis is an unresolved risk after coronary intervention. Biodegradable stents would provide initial scaffolding of the stenosed segment and disappear subsequently. The additive manufacturing technology Selective Laser Melting (SLM) enables rapid, parallel, and raw material saving generation of complex 3- dimensional structures with extensive geometric freedom and is currently in use in orthopedic or dental applications. Here, SLM process parameters were adapted for poly-L-lactid acid (PLLA) and PLLA-co-poly-ε-caprolactone (PCL) powders to generate degradable coronary stent prototypes. Biocompatibility of both polymers was evidenced by assessment of cell morphology and of metabolic and adhesive activity at direct and indirect contact with human coronary artery smooth muscle cells, umbilical vein endothelial cells, and endothelial progenitor cells. γ-sterilization was demonstrated to guarantee safety of SLM-processed parts. From PLLA and PCL, stent prototypes were successfully generated and post-processing by spray- and dip-coating proved to thoroughly smoothen stent surfaces. In conclusion, for the first time, biodegradable polymers and the SLM technique were combined for the manufacturing of customized biodegradable coronary artery stent prototypes. SLM is advocated for the development of biodegradable coronary PLLA and PCL stents, potentially optimized for future bifurcation applications.
Collapse
Affiliation(s)
- Christian Flege
- Department of Cardiology, RWTH Aachen University, Aachen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Percutaneous transluminal coronary angioplasty is a widely used technique for recanalizing arteries that are occluded by atherosclerotic plaque, but its usefulness is limited by the occurrence ofrestenosis in a high proportion of patients. The development of new therapies for this currently intractable problem will be facilitated by the use of animal models of restenosis that are predictive of drug efficacy in humans. Two approaches for improving predictivity can be identified. In the first of these, the goal is to maximize the anatomical and procedural resemblance of the model to humans. The second approach seeks to maximize the pathophysiological and molecular biological resemblance of the model to humans. Tangible progress is being made toward the first goal, but lack of understanding of the basic biology of human restenosis is hampering progress toward the second.
Collapse
|
39
|
The attenuation of restenosis following arterial gene transfer using carbon nanotube coated stent incorporating TAT/DNAAng1+Vegf nanoparticles. Biomaterials 2012; 33:7655-64. [DOI: 10.1016/j.biomaterials.2012.06.096] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 06/29/2012] [Indexed: 11/20/2022]
|
40
|
Pettersen RJ, Salem M, Rotevatn S, Kuiper KK, Larsen TH, Bohov P, Berge RK, Nordrehaug JE. Effects of local delivery of Tetradecylthioacetic acid within the injured coronary vessel wall. SCAND CARDIOVASC J 2012; 46:366-73. [DOI: 10.3109/14017431.2012.725477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
41
|
Coronary angiography and percutaneous coronary intervention in the porcine model: a practical guide to the procedure. Animal 2012; 6:311-20. [PMID: 22436190 DOI: 10.1017/s1751731111001650] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Assessment of safety and efficacy within the porcine coronary artery model remains a standard requirement for new therapies delivered to the coronary arteries before proceeding to clinical testing. Human coronary procedures carry a very low mortality rate; however, procedural mortality for porcine experiments is often high, despite these animals being young and free of atherosclerosis. Some of these deaths are due to poor technique, and therefore avoidable. However, despite the wide use of this model, a systematic description of the procedure has never been published. This article will detail how porcine angiography and stent implantation is performed in our institution and will discuss the relevant differences between humans and pigs with regard to anaesthesia, pharmacotherapy, vascular access, catheter selection and angiographic views. Important variations to the technique that have been reported are also covered.
Collapse
|
42
|
Gupta GK, Agrawal T, Del Core MG, Hunter WJ, Agrawal DK. Decreased expression of vitamin D receptors in neointimal lesions following coronary artery angioplasty in atherosclerotic swine. PLoS One 2012; 7:e42789. [PMID: 22880111 PMCID: PMC3412822 DOI: 10.1371/journal.pone.0042789] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 07/12/2012] [Indexed: 11/18/2022] Open
Abstract
Background Inflammatory cytokines, such as TNF-α, play a key role in the pathogenesis of occlusive vascular diseases. Activation of vitamin D receptors (VDR) elicits both growth-inhibitory and anti-inflammatory effects. Here, we investigated the expression of TNF-α and VDR in post-angioplasty coronary artery neointimal lesions of hypercholesterolemic swine and examined the effect of vitamin D deficiency on the development of coronary restenosis. We also examined the effect of calcitriol on cell proliferation and effect of TNF-α on VDR activity and expression in porcine coronary artery smooth muscle cells (PCASMCs) in-vitro. Methodology/Principal Findings Expression of VDR and TNF-α and the effect of vitamin D deficiency in post-angioplasty coronary arteries were analyzed by immunohistochemistry and histomorphometry. Cell proliferation was examined by thymidine and BrdU incorporation assays in cultured PCASMCs. Effect of TNF-α-stimulation on the activity and expression of VDR was analyzed by luciferase assay, immunoblotting and immunocytochemistry. In-vivo, morphometric analysis of the tissues revealed typical lesions with significant neointimal proliferation. Histological evaluation showed expression of smooth muscle α-actin and significantly increased expression of TNF-α in neointimal lesions. Interestingly, there was significantly decreased expression of VDR in PCASMCs of neointimal region compared to normal media. Indeed, post-balloon angioplasty restenosis was significantly higher in vitamin D-deficient hypercholesterolemic swine compared to vitamin D-sufficient group. In-vitro, calcitriol inhibited both serum- and PDGF-BB-induced proliferation in PCASMCs and TNF-α-stimulation significantly decreased the expression and activity of VDR in PCASMCs. Conclusions/Significance These data suggest that significant downregulation of VDR in proliferating smooth muscle cells in neointimal lesions could be due to atherogenic cytokines, including TNF-α. Vitamin D deficiency potentiates the development of coronary restenosis. Calcitriol has anti-proliferative properties in PCASMCs and these actions are mediated through VDR. This could be a potential mechanism for uncontrolled growth of neointimal cells in injured arteries leading to restenosis.
Collapse
Affiliation(s)
- Gaurav K. Gupta
- Department of Biomedical Sciences and Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, United States of America
| | - Tanupriya Agrawal
- Department of Biomedical Sciences and Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, United States of America
| | - Michael G. Del Core
- Department of Internal Medicine, Creighton University School of Medicine, Omaha, Nebraska, United States of America
| | - William J. Hunter
- Department of Pathology, Creighton University School of Medicine, Omaha, Nebraska, United States of America
| | - Devendra K. Agrawal
- Department of Biomedical Sciences and Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, United States of America
- Department of Internal Medicine, Creighton University School of Medicine, Omaha, Nebraska, United States of America
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
43
|
Kondyurin AV, Maitz MF, Romanova VA, Begishev VP, Kondyurina IV, Guenzel R. Drug release from polyureaurethane coating modified by plasma immersion ion implantation. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2012; 15:145-59. [PMID: 15109094 DOI: 10.1163/156856204322793548] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A crosslinked polyurethanurea (PUU) coating was synthesised from a solution on metal vascular stents. In the model system the glucocorticoid prednisolone was inserted into the film by the equilibrium swelling method; after this plasma immersion ion implantation (PIII) was applied to modify the coating for improved release kinetics. This treatment causes the formation of oxygen-containing and unsaturated carbon-carbon groups in the PUU and a destruction of the drug in the surface layer. As a consequence, the release rate of prednisolone to water becomes more stable with time than it is at the untreated coating. In this drug release system PIII treatment prevents an initial toxically high release of the drug. By this it allows the incorporation of a higher amount of the drug and an extended action.
Collapse
Affiliation(s)
- A V Kondyurin
- Institute of Polymer Research, Dresden 01069, Germany.
| | | | | | | | | | | |
Collapse
|
44
|
Fefer P, Robert N, Qiang B, Liu G, Munce N, Anderson K, Osherov A, Ladouceur-Wodzak M, Qi X, Dick A, Weisbrod M, Samuel M, Butany J, Wright G, Strauss B. Characterisation of a novel porcine coronary artery CTO model. EUROINTERVENTION 2012; 7:1444-52. [DOI: 10.4244/eijv7i12a225] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
45
|
Houbballah R, Robaldo A, Albadawi H, Titus J, LaMuraglia GM. A novel model of accelerated intimal hyperplasia in the pig iliac artery. Int J Exp Pathol 2011; 92:422-7. [PMID: 22050434 DOI: 10.1111/j.1365-2613.2011.00790.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
There is no good animal model of large artery injury-induced intimal hyperplasia (IH). Those available are reproducible, providing only a few layers of proliferating cells or have the disadvantage of the presence of a metallic stent that complicates histology evaluation. This study was designed to develop a new, simple model of accelerated IH based on balloon injury in conjunction with disruption of the Internal Elastic Lamina (IEL) in pig external iliac arteries. Iliac artery injury (n = 24) was performed in 12 Yorkshire pigs divided in two groups: Group I (n = 10), overdistention injury induced by an oversized non-compliant balloon; Group II (n = 14), arterial wall disruption by pulling back an isometric cutting balloon (CB) followed by stretching with a compliant Fogarty Balloon (FB). At two weeks, arteries were processed for morphometric analysis and immunohistochemistry (IHC) for smooth muscle cells (SMC) and proliferating cell nuclear antigen (PCNA). When comparing the two groups, at 2 weeks, arteries of group II had a higher incidence of IH (100%vs. 50%, P = 0.0059), increased intimal areas (2.54 ± 0.33 mm(2) vs. 0.93 ± 0.36 mm(2) , P = 0.004), increased intimal area/Media area ratios (0.95 ± 0.1 vs. 0.28 ± 0.05; P < 0.0001) and decreased lumen areas (6.24 ± 0.44 vs. 9.48 ± 1.56, P = 0.026). No thrombosis was noticed in Group II. Neointima was composed by proliferating SMC located with the highest concentration in the area of IEL disruption (IHC). Arterial injury by pulling back CB and FB induces significant IH in pig iliac arteries by two weeks without thrombosis. This model is superior to the classical overdistention non-compliant model and should be useful and cost-effective for preclinical testing of procedures designed to inhibit IH in large peripheral arteries.
Collapse
Affiliation(s)
- Rabih Houbballah
- Division of Vascular and Endovascular Surgery of the General Surgical Services, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | | | | | | | | |
Collapse
|
46
|
Lim S, Yoon JW, Kang SM, Choi SH, Cho BJ, Kim M, Park HS, Cho HJ, Shin H, Kim YB, Kim HS, Jang HC, Park KS. EGb761, a Ginkgo biloba extract, is effective against atherosclerosis in vitro, and in a rat model of type 2 diabetes. PLoS One 2011; 6:e20301. [PMID: 21655098 PMCID: PMC3107221 DOI: 10.1371/journal.pone.0020301] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Accepted: 04/29/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND EGb761, a standardized Ginkgo biloba extract, has antioxidant and antiplatelet aggregation and thus might protect against atherosclerosis. However, molecular and functional properties of EGb761 and its major subcomponents have not been well characterized. We investigated the effect of EGb761 and its major subcomponents (bilobalide, kaemferol, and quercetin) on preventing atherosclerosis in vitro, and in a rat model of type 2 diabetes. METHODS AND RESULTS EGb761 (100 and 200 mg/kg) or normal saline (control) were administered to Otsuka Long-Evans Tokushima Fatty rats, an obese insulin-resistant rat model, for 6 weeks (from 3 weeks before to 3 weeks after carotid artery injury). Immunohistochemical staining was performed to investigate cell proliferation and apoptosis in the injured arteries. Cell migration, caspase-3 activity and DNA fragmentation, monocyte adhesion, and ICAM-1/VCAM-1 levels were explored in vitro. Treatment with EGb761 dose-dependently reduced intima-media ratio, proliferation of vascular smooth muscle cells (VSMCs) and induced greater apoptosis than the controls. Proliferation and migration of VSMCs in vitro were also decreased by the treatment of EGb761. Glucose homeostasis and circulating adiponectin levels were improved, and plasma hsCRP concentrations were decreased in the treatment groups. Caspase-3 activity and DNA fragmentation increased while monocyte adhesion and ICAM-1/VCAM-1 levels decreased significantly. Among subcomponents of EGb761, kaemferol and quercetin reduced VSMC migration and increased caspase activity. CONCLUSIONS EGb761 has a protective role in the development of atherosclerosis and is a potential therapeutic agent for preventing atherosclerosis.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Atherosclerosis/prevention & control
- Caspase 3/metabolism
- Cell Adhesion/drug effects
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Cyclopentanes/pharmacology
- Diabetes Mellitus, Type 2/prevention & control
- Disease Models, Animal
- Furans/pharmacology
- Ginkgo biloba/chemistry
- Ginkgolides/pharmacology
- Humans
- Immunoblotting
- Kaempferols/pharmacology
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Plant Extracts/pharmacology
- Quercetin/pharmacology
- Rats
- Rats, Inbred OLETF
- Reverse Transcriptase Polymerase Chain Reaction
- Tunica Intima/drug effects
- Tunica Intima/metabolism
- Tunica Intima/pathology
- U937 Cells
Collapse
Affiliation(s)
- Soo Lim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Ji Won Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Seon Mee Kang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Sung Hee Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Bong Jun Cho
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Min Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Ho Seon Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Hyun Ju Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Hayley Shin
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hyo Soo Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Hak Chul Jang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kyong Soo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
- * E-mail:
| |
Collapse
|
47
|
Timmins LH, Miller MW, Clubb FJ, Moore JE. Increased artery wall stress post-stenting leads to greater intimal thickening. J Transl Med 2011; 91:955-67. [PMID: 21445059 PMCID: PMC3103652 DOI: 10.1038/labinvest.2011.57] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Since the first human procedure in the late 1980s, vascular stent implantation has been accepted as a standard form of treatment of atherosclerosis. Despite their tremendous success, these medical devices are not without their problems, as excessive neointimal hyperplasia can result in the formation of a new blockage (restenosis). Clinical data suggest that stent design is a key factor in the development of restenosis. Additionally, computational studies indicate that the biomechanical environment is strongly dependent on the geometrical configuration of the stent, and, therefore, possibly involved in the development of restenosis. We hypothesize that stents that induce higher stresses on the artery wall lead to a more aggressive pathobiologic response, as determined by the amount of neointimal hyperplasia. The aim of this investigation was to examine the role of solid biomechanics in the development of restenosis. A combination of computational modeling techniques and in vivo analysis were employed to investigate the pathobiologic response to two stent designs that impose greater or lesser levels of stress on the artery wall. Stent designs were implanted in a porcine model (pigs) for approximately 28 days and novel integrative pathology techniques (quantitative micro-computed tomography, histomorphometry) were utilized to quantify the pathobiologic response. Concomitantly, computational methods were used to quantify the mechanical loads that the two stents place on the artery. Results reveal a strong correlation between the computed stress values induced on the artery wall and the pathobiologic response; the stent that subjected the artery to the higher stresses had significantly more neointimal thickening at stent struts (high-stress stent: 0.197±0.020 mm vs low-stress stent: 0.071±0.016 mm). Therefore, we conclude that the pathobiologic differences are a direct result of the solid biomechanical environment, confirming the hypothesis that stents that impose higher wall stresses will provoke a more aggressive pathobiological response.
Collapse
Affiliation(s)
- Lucas H. Timmins
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA 77843-3120
| | - Matthew W. Miller
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA 77843-4474
- M. E. DeBakey Institute, Texas A&M University, College Station, TX, USA
| | - Fred J. Clubb
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, USA 77843-4467
- M. E. DeBakey Institute, Texas A&M University, College Station, TX, USA
| | - James E. Moore
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA 77843-3120
- M. E. DeBakey Institute, Texas A&M University, College Station, TX, USA
| |
Collapse
|
48
|
Violaris A, Francis S, Holt C, Clelland C, Gadsdon P, Angelini G. Organ culture of human coronary artery following balloon angioplasty. Int J Angiol 2011. [DOI: 10.1007/bf02651568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
49
|
Milewski K, Zurakowski A, Pajak J, Liszka L, Debinski M, Buszman PP, Samra MA, Dominek P, Aboodi MS, Kaluza GL, Buszman P. Effects of local intracoronary paclitaxel delivery using the Remedy transport catheter on neointimal hyperplasia after stent implantation in a porcine model. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2011; 12:82-9. [DOI: 10.1016/j.carrev.2010.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2010] [Revised: 04/30/2010] [Accepted: 05/11/2010] [Indexed: 10/18/2022]
|
50
|
Lim S, Moon MK, Shin H, Kim TH, Cho BJ, Kim M, Park HS, Choi SH, Ko SH, Chung MH, Lee IK, Jang HC, Kim YB, Park KS. Effect of S-adenosylmethionine on neointimal formation after balloon injury in obese diabetic rats. Cardiovasc Res 2011; 90:383-93. [PMID: 21245056 DOI: 10.1093/cvr/cvr009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
AIMS The association between hyperhomocysteinaemia and cardiovascular disease has been attributed to low levels of S-adenosylmethionine (SAM), a metabolic intermediate of homocysteine. However, the role of SAM in the development of restenosis has not been explored. Therefore, we investigated the effects of SAM on neointimal formation after balloon injury in obese diabetic rats and cultured cells. METHODS AND RESULTS Otsuka Long-Evans Tokushima fatty rats were divided into the following three groups: control (normal saline); SAM15; and SAM30 (15 and 30 mg/kg per day, respectively; n = 10 per group). SAM was administered orally from 1 week before carotid injury to 2 weeks thereafter. SAM treatment for 3 weeks caused a significant dose-dependent reduction in the intima-to-media ratio. SAM treatment significantly reduced the proliferation of vascular smooth muscle cells (VSMCs) and induced more apoptosis than was observed in the control group. This effect was accompanied by reduced circulating levels of high-sensitivity C-reactive protein and monocyte chemoattractant protein-1, reduced urine 8-hydroxy-2'-deoxyguanosine (8-OHdG), and increased adiponectin. Intima-to-media ratio correlated significantly with the levels of inflammatory markers, adiponectin, and 8-OHdG. In vitro experiments demonstrated that VSMC proliferation and migration and the adhesion of monocytes decreased in response to SAM. SAM treatment also reduced tumour necrosis factor-α-induced reactive oxygen species and tunicamycin-induced GRP78 expression in VSMCs. CONCLUSION These findings suggest that SAM exerts protective effects against restenosis after balloon injury in a rat model of type 2 diabetes by reducing the proliferation and inducing the apoptosis of VSMCs, modifying the inflammatory processes and reducing oxidative and endoplasmic reticulum stresses.
Collapse
Affiliation(s)
- Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine, 28 Yongon-Dong, Chongno-Gu, Seoul, Korea 110-744
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|