1
|
Oh K, Repasky A, Nader ND, Rivero M, Montross B, Khan SZ, Harris L, Dryjski M, Dosluoglu HH. Hemoglobin A1c levels are related to patency and adverse limb events in diabetics after revascularization. J Vasc Surg 2024:S0741-5214(24)01881-0. [PMID: 39303863 DOI: 10.1016/j.jvs.2024.09.003] [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: 06/10/2024] [Revised: 08/27/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVE Poor glycemic control in the perioperative period has been reported to be associated with early and late major adverse limb events (MALEs). However, these studies were mostly from large databases or lacked long-term outcomes. We examined the long-term effects of high hemoglobin A1c (HbA1c) level on patency, MALE, limb salvage (LS) and survival after lower extremity revascularization procedures in patients with diabetes. METHODS Patients with diabetes who underwent revascularization for Rutherford class 3 to 6 ischemia between May 2002 and December 2018 were identified. Patients with an HbA1c of ≤7% were compared with those with an HbA1c of >7% for patency, MALE, survival, LS, and amputation-free survival. RESULTS Of 706 patients, 699 had HbA1c data (775 limbs), with 311 (357 limbs) in the HbA1c ≤7% and 388 (418 limbs) in the HbA1c >7% groups. Patients with an HbA1c of >7% were younger (69.9 ± 10.2 years vs 71.7 ± 9.5 years; P = .011), had higher lipid levels, insulin use (70% vs 49%; P < .01), American Society of Anesthesiologists classification of 4, and had a lower prevalence of chronic kidney disease (32% v s41%; P = .023). Patients with an HbA1c of >7% were more likely to present with chronic limb-threatening ischemia (CLTI) (79% vs 72%; P = .019) and undergo infrapopliteal interventions (49% vs 42%; P = .005), with no difference in anatomical complexity (TransAtlantic Inter Society Consensus class C/D, 75% vs 77%; P = .72) or type of revascularization (24% vs 18% open, 66% vs 70% endovascular, 10% vs 12% hybrid; P = .236). Patency and freedom from MALE were significantly lower in patients with an HbA1c of >7% for infrainguinal revascularizations, whereas amputation-free survival and overall survival were similar. In patients with chronic limb-threatening ischemia, LS rates at 5 years were significantly lower in patients undergoing open revascularization (HbA1c > 7%: 64% ± 6% vs HbA1c < 7%:86% ± 5%; P = .020), whereas it was similar after endovascular interventions (HbA1c > 7%:79% ± 4% vs HbA1c < 7%:77% ± 3%; P = .631). Seventy patients with an HbA1c of >7% lost limbs vs 38 patients with an HbA1c of ≤7% (P = .007). In multivariate analysis, HbA1c was associated significantly with primary patency. HbA1c, insulin use, level of intervention, and angiotensin-converting enzyme inhibitor use were associated with MALE. CONCLUSIONS A perioperative HbA1c of >7% is associated with poorer patency rates and increased MALE, especially at the infrainguinal level revascularization in patients with diabetes, with no significant impact on survival. LS is impacted after open, but not after endovascular revascularization.
Collapse
Affiliation(s)
- Kenny Oh
- Division of Vascular Surgery, Department of Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA.
| | - Amber Repasky
- Department of Anesthesiology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY
| | - Nader D Nader
- Department of Anesthesiology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY
| | - Mariel Rivero
- Division of Vascular Surgery, Department of Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Division of Vascular Surgery, VA Western NY Healthcare System, Buffalo, NY
| | - Brittany Montross
- Division of Vascular Surgery, Department of Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Sikandar Z Khan
- Division of Vascular Surgery, Department of Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Linda Harris
- Division of Vascular Surgery, Department of Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Maciej Dryjski
- Division of Vascular Surgery, Department of Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Hasan H Dosluoglu
- Division of Vascular Surgery, Department of Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Division of Vascular Surgery, VA Western NY Healthcare System, Buffalo, NY
| |
Collapse
|
2
|
Yap T, Silickas J, Weerakkody R, Lea T, Santhirakumaran G, Bremner L, Diamantopoulos A, Biasi L, Thomas S, Zayed H, Patel SD. Predictors of outcome in diabetic patients undergoing infrapopliteal endovascular revascularization for chronic limb-threatening ischemia. J Vasc Surg 2021; 75:618-624. [PMID: 34634414 DOI: 10.1016/j.jvs.2021.09.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/22/2021] [Indexed: 12/01/2022]
Abstract
OBJECTIVE The incidence of chronic limb-threatening ischemia in diabetic patients is increasing. The factors influencing outcome after infrapopliteal revascularization in these patients are largely unknown. Therefore, this study aims to identify the impact of perioperative glucose control on the long-term outcomes in this patient cohort, and furthermore to identify other factors independently associated with outcome. METHODS Consecutive diabetic patients undergoing infrapopliteal endovascular revascularization for chronic limb-threatening ischemia were identified. Patients' demographics, procedural details, daily capillary blood glucose, and hemoglobin A1C levels were collected and analyzed against the study end points using Kaplan-Meier and Cox regression analysis. RESULTS A total of 437 infrapopliteal target vessels were successfully crossed in 203 patients. Amputation-free survival by Kaplan-Meier (estimate (standard error)%) was 74 (3.3)% and 63 (3.7)%, primary patency was 61 (4.2)% and 50 (4.9)%, assisted primary patency was 69 (5.2)% and 55 (6.1)%, and secondary patency was 71 (3.8)% and 59 (4.1)% at 1 year and 2 years, respectively. Cox regression analysis showed high perioperative capillary blood glucose levels to be an independent predictor of binary restenosis (hazard ratio [HR], 1.49; 95% confidence interval [CI], 1.31-1.1.78; P = .015). Postprocedural dual-antiplatelet therapy was found to be an independent predictor of amputation-free survival (HR, 1.69; 95% CI, 1.04-2.75; P = .033), and freedom from major adverse limb events (HR: 1.96; 95% CI, 1.16-3.27; P = .023) and baseline estimated glomerular filtration rate was significantly associated with better amputation-free survival (HR, 0.52; 95% CI, 0.31-0.87; P = .014). CONCLUSIONS Poor perioperative glycemic control is associated with a higher incidence of restenosis after infrapopliteal revascularization in diabetic patients. Dual antiplatelet therapy is associated with better outcomes in this group.
Collapse
Affiliation(s)
- Trixie Yap
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom.
| | - Justinas Silickas
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom
| | - Ruwan Weerakkody
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom
| | - Talia Lea
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom
| | - Gowthanan Santhirakumaran
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom
| | - Laura Bremner
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom
| | - Athanasios Diamantopoulos
- Department of Interventional Radiology, Guys' and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Lukla Biasi
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom
| | - Stephen Thomas
- Department of Endocrinology and Metabolic Medicine, Guys' and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Hany Zayed
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom
| | - Sanjay D Patel
- Department of Vascular & Endovascular Surgery, Guy's and St Thomas NHS Foundation Trust, King's Health Partners, London, United Kingdom
| |
Collapse
|
3
|
Kim J, Park TK, Choi KH, Choi D, Ko YG, Lee JH, Yoon CH, Chae IH, Yu CW, Min PK, Lee SW, Lee SR, Koh YS, Choi SH. Different association between renal dysfunction and clinical outcomes according to the presence of diabetes in patients undergoing endovascular treatment for peripheral artery disease. J Vasc Surg 2019; 71:132-140.e1. [PMID: 31285068 DOI: 10.1016/j.jvs.2019.03.071] [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: 12/18/2018] [Accepted: 03/30/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Although chronic kidney disease (CKD) and diabetes are important prognostic factors in patients with peripheral artery disease, there are limited data regarding the outcomes of endovascular treatment (EVT) according to the severity of CKD, especially in the presence of diabetes. This study sought to compare clinical outcomes of lower limb EVT between patients with and patients without CKD according to the presence of diabetes. METHODS Patients were enrolled from the Korean multicenter EVT registry and were divided according to the presence of diabetes, then further stratified by CKD (estimated glomerular filtration rate <60 mL/min/1.73 m2). The primary outcome was major adverse limb events (MALEs; a composite of reintervention for target limb, reintervention for target vessel, and unplanned major amputation) at 2 years. RESULTS A total of 3045 patients were eligible for analysis: 1277 nondiabetic patients (944 without CKD, 333 with CKD) and 1768 diabetic patients (951 without CKD, 817 with CKD). CKD was associated with a significantly increased risk of MALEs after EVT in diabetic patients (14.4% vs 9.9%; adjusted hazard ratio, 1.60; 95% confidence interval, 1.28-2.01; P < .001) but not in nondiabetic patients (7.6% vs 9.7%; adjusted hazard ratio, 0.78; 95% confidence interval, 0.53-1.14; P = .203; interaction P = .018). In analysis stratified by the severity of CKD among diabetic patients, end-stage renal disease was significantly associated with an increased risk of MALE. CONCLUSIONS CKD was associated with a significantly higher risk of MALEs after EVT in diabetic patients but not in nondiabetic patients. The increased risk of MALEs was mainly driven by patients with end-stage renal disease.
Collapse
Affiliation(s)
- Jihoon Kim
- Division of Cardiology, Department of Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Taek Kyu Park
- Division of Cardiology, Department of Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ki Hong Choi
- Division of Cardiology, Department of Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Donghoon Choi
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young-Guk Ko
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-Hwan Lee
- Division of Cardiology, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Chang-Hwan Yoon
- Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - In-Ho Chae
- Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Cheol Woong Yu
- Division of Cardiology, Department of Internal Medicine, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Pil-Ki Min
- Division of Cardiology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung-Whan Lee
- Division of Cardiology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang-Rok Lee
- Division of Cardiology, Department of Internal Medicine, Chonbuk National University Hospital, Seoul, Republic of Korea
| | - Yoon Seok Koh
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung-Hyuk Choi
- Division of Cardiology, Department of Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | | |
Collapse
|
4
|
Kuroda M, Shinke T, Otake H, Sugiyama D, Takaya T, Takahashi H, Terashita D, Uzu K, Tahara N, Kashiwagi D, Kuroda K, Shinkura Y, Nagasawa Y, Sakaguchi K, Hirota Y, Ogawa W, Hirata KI. Effects of daily glucose fluctuations on the healing response to everolimus-eluting stent implantation as assessed using continuous glucose monitoring and optical coherence tomography. Cardiovasc Diabetol 2016; 15:79. [PMID: 27208906 PMCID: PMC4875633 DOI: 10.1186/s12933-016-0395-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/10/2016] [Indexed: 11/30/2022] Open
Abstract
Background Several studies have revealed that glucose fluctuations provoke oxidative stress that leads to endothelial cell dysfunction, progression of coronary atherosclerosis, and plaque vulnerability. However, little is known regarding their effect on neointimal growth after stenting in patients with coronary artery disease (CAD). We aimed to investigate the effects of glucose fluctuations on neointimal growth after everolimus-eluting stent (EES) implantation. Methods This study examined 50 patients who underwent a 9-month follow-up using optical coherence tomography (OCT) after EES implantation. Glucose fluctuation was expressed as the mean amplitude of glycemic excursion (MAGE), and was determined via continuous glucose monitoring before stenting. At the OCT follow-up, we evaluated the percentage of uncovered struts and three-dimensional uniformity of neointimal distribution by calculating the mean neointimal thickness (NIT) within 360 equally-spaced radial sectors for every 1-mm cross-sectional OCT analysis, and assessed the incidence of major adverse cardiovascular events (MACE). Results We evaluated 60 lesions in 50 patients. Linear mixed effect models were used to explore the influence of different variables on variability in NIT and the percentage of uncovered struts and to adjust for covariates. Univariate analysis showed that MAGE was most strongly correlated with the previously mentioned OCT measurements (coefficient β ± standard error = 0.267 ± 0.073 and 0.016 ± 0.003, t = 3.668 and 6.092, both P < 0.001, respectively). In multivariate analysis, MAGE had the strongest effect on variability in NIT (coefficient β ± standard error = 0.239 ± 0.093, P = 0.014) and the percentage of uncovered struts (coefficient β ± standard error = 0.019 ± 0.004, P < 0.001). Five lesions in four patients required target lesion revascularization (10.0 %) at a mean duration of 9 months after EES implantation. Compared to non-MACE cases, cases of MACE exhibited a significantly higher MAGE (99 vs. 68; P = 0.004), maximum NIT (580 vs. 330 µm; P = 0.002), and variability in NIT (100 vs. 65; P = 0.007), although there was no significant difference in these groups’ HbA1c levels. Conclusions Glucose fluctuation may affect vessel healing after EES implantation in patients with CAD who are receiving lipid-lowering therapy. Therefore, glucose fluctuations may be an important target for secondary prevention after coronary stenting, which is independent of dyslipidemia control.
Collapse
Affiliation(s)
- Masaru Kuroda
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Toshiro Shinke
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan.
| | - Hiromasa Otake
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Daisuke Sugiyama
- Department of Preventive Medicine and Public Health, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Tomofumi Takaya
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Hachidai Takahashi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Daisuke Terashita
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Kenzo Uzu
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Natsuko Tahara
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Daiji Kashiwagi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Koji Kuroda
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Yuto Shinkura
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Yoshinori Nagasawa
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Kazuhiko Sakaguchi
- Division of Diabetes and Metabolism, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Yushi Hirota
- Division of Diabetes and Metabolism, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Wataru Ogawa
- Division of Diabetes and Metabolism, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| |
Collapse
|
5
|
Fu Y, Duru EA, Davies MG. Effect of metabolic syndrome on the response to arterial injury. J Surg Res 2014; 191:33-41. [PMID: 24972735 DOI: 10.1016/j.jss.2014.05.051] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 04/27/2014] [Accepted: 05/16/2014] [Indexed: 12/01/2022]
Abstract
BACKGROUND Metabolic syndrome is now an epidemic in the United States population. Intimal hyperplasia remains the principal lesion in the development of restenosis after vessel wall injury. The aim of this study is to characterize the changes induced in wall morphology in the developing intimal hyperplasia within a murine model in the presence of diabetes (type 1) and metabolic syndrome. METHODS Control (wild type B6), Non Obese Diabetic, and metabolic syndrome (RCS-10) mice were used. The murine femoral wire injury model was used in which a micro wire is passed through a branch of the femoral and used to denude the common femoral and iliac arteries. Specimens were perfusion fixed and sections were stained with hematoxylin and eosin and Movat stains such that dimensional and compositional morphometry could be performed using an ImagePro system. Additional stains for proliferation and apoptosis were used. RESULTS In control mice, the injured femoral arteries develop intimal hyperplasia, which is maximal at 28 d and remains stable to day 56. Sham-operated vessels do not produce such a response. In diabetic mice, the intimal response increased 5-fold with a 2-fold increase in proteoglycan deposition, whereas in the metabolic syndrome mice there was a 6-fold increase in the intimal response and a similar increase in proteoglycan deposition. Collagen deposition was different with a 22-fold increase over control in collagen deposition in diabetes and a 100-fold increase over control in collagen deposition in metabolic syndrome as compared with the control injury mice. Maximal vascular smooth muscle cell (VSMC) proliferation was decreased in both diabetes and metabolic syndrome compared with controls, whereas early cell apoptosis in both diabetes and metabolic syndrome was sustained over a longer period of time compared with wild-type mice. CONCLUSIONS These data demonstrate that development of intimal hyperplasia is markedly different in diabetes and metabolic syndrome compared with controls, with an increase in collagen deposition, a reduction in VSMC proliferation, and an increase in early VSMC apoptosis. These findings suggest that preventative strategies against restenosis must be tailored for the diabetic and metabolic syndrome patients.
Collapse
Affiliation(s)
- Yuyang Fu
- Vascular Biology and Therapeutics Program, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas; Department of Cardiovascular Surgery, Houston Methodist DeBakey Heart & Vascular Center, Houston Methodist Hospital, Houston, Texas
| | - Enrico A Duru
- Vascular Biology and Therapeutics Program, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas; Department of Cardiovascular Surgery, Houston Methodist DeBakey Heart & Vascular Center, Houston Methodist Hospital, Houston, Texas
| | - Mark G Davies
- Vascular Biology and Therapeutics Program, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas; Department of Cardiovascular Surgery, Houston Methodist DeBakey Heart & Vascular Center, Houston Methodist Hospital, Houston, Texas.
| |
Collapse
|
6
|
Tanaka N, Terashima M, Rathore S, Itoh T, Habara M, Nasu K, Kimura M, Itoh T, Kinoshita Y, Ehara M, Tsuchikane E, Asakura K, Asakura Y, Katoh O, Suzuki T. Different patterns of vascular response between patients with or without diabetes mellitus after drug-eluting stent implantation: optical coherence tomographic analysis. JACC Cardiovasc Interv 2011; 3:1074-9. [PMID: 20965467 DOI: 10.1016/j.jcin.2010.08.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Revised: 06/17/2010] [Accepted: 08/05/2010] [Indexed: 10/18/2022]
Abstract
OBJECTIVES We performed this study to investigate with optical coherence tomography (OCT) the vascular response after sirolimus-eluting stent (SES) implantation between patients with and those without diabetes mellitus (DM). BACKGROUND The difference in vascular response after SES implantation between patients with and those without DM has not been fully evaluated with OCT. METHODS Optical coherence tomography was performed to examine 74 nonrestenotic SES implanted in 63 patients (32 with DM and 31 without DM) at 9 months after SES implantation. For struts showing neointimal coverage, the neointimal thickness on the luminal side of each strut section was measured, and neointimal characteristics were classified into high, low, and layered signal pattern. RESULTS Baseline patient characteristics and lesion and procedural characteristics data were similar between the 2 groups. In total, 11,422 struts were analyzed. High signal neointima was observed in 90.2 ± 13.9%, low signal neointima in 7.3 ± 10.0%, and layered neointima in 2.7 ± 5.8%/stents. There was higher incidence of low signal neointima (10.5 ± 10.3% vs. 4.5 ± 5.6%, p = 0.003), neointimal thickness was larger (median: 106.8 μm, interquartile range: 79.3 to 130.4 μm vs. median: 83.5 μm, interquartile range: 62.3 to 89.3 μm; p < 0.0001), and neointimal coverage of stent struts was higher (92.1 ± 6.2% vs. 87.2 ± 11.9%; p = 0.03) in DM patients. CONCLUSIONS High signal neointimal pattern was predominantly observed, and low or layered signal pattern was observed in some cases. In DM patients, low signal neointima was observed with high frequency. Neointimal coverage and neointimal thickness was also higher in DM patients as compared with non-DM patients.
Collapse
Affiliation(s)
- Nobuyoshi Tanaka
- Department of Cardiology, Toyohashi Heart Center, Toyohashi, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Takahashi M, Bujo H, Jiang M, Noike H, Saito Y, Shirai K. Enhanced circulating soluble LR11 in patients with coronary organic stenosis. Atherosclerosis 2010; 210:581-4. [DOI: 10.1016/j.atherosclerosis.2009.12.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Revised: 11/27/2009] [Accepted: 12/07/2009] [Indexed: 11/17/2022]
|
8
|
Ng VG, Stone GW. Diabetics and drug-eluting stents in ST segment elevation myocardial infarction: confidence in numbers. Rev Esp Cardiol 2009; 62:343-6. [PMID: 19401117 DOI: 10.1016/s1885-5857(09)71659-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
9
|
Diabéticos y stents liberadores de fármacos en el infarto de miocardio con elevación del segmento ST: confianza en las cifras. Rev Esp Cardiol 2009. [DOI: 10.1016/s0300-8932(09)70889-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
10
|
Akiyama N, Naruse K, Kobayashi Y, Nakamura N, Hamada Y, Nakashima E, Matsubara T, Oiso Y, Nakamura J. High glucose-induced upregulation of Rho/Rho-kinase via platelet-derived growth factor receptor-beta increases migration of aortic smooth muscle cells. J Mol Cell Cardiol 2008; 45:326-32. [PMID: 18561944 DOI: 10.1016/j.yjmcc.2008.04.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Revised: 03/25/2008] [Accepted: 04/10/2008] [Indexed: 11/17/2022]
Abstract
Small GTPase Rho and Rho-kinase, the target protein of Rho, play an important role in atherosclerosis. In diabetic macroangiopathy, one of the major pathogenic changes is the migration of vascular smooth muscle cells (SMCs). Platelet-derived growth factor (PDGF) is known to stimulate the migration of SMCs. In the current study, we have investigated the involvement of the Rho/Rho-kinase pathway in the increased migration of cultured human aortic SMCs under a high glucose condition. PDGF stimulated the activation and the protein level of Rho. The protein level of PDGF receptor-beta (PDGFR-beta) was increased under the high glucose condition concomitant with the increased protein level and activation of Rho. The increased protein level and activity of Rho were suppressed by an anti-PDGF neutralizing antibody or a PDGFR-beta inhibitor, AG1433, under the high glucose condition. Furthermore, high glucose significantly increased the migration of SMCs. A specific inhibitor of Rho-kinase, Y-27632, or anti-PDGF neutralizing antibody inhibited increased migration of SMCs under the high glucose condition. The protein levels of Rho were increased in aortae of diabetic rats, which were abolished by the treatment of Imatinib, the inhibitor of PDGFR. These observations indicate that the upregulation of the PDGFR-beta / Rho / Rho-kinase pathway increases the migration of SMCs under the high glucose condition. The inhibition of Rho/Rho-kinase may be a new target for the treatment of diabetic macroangiopathy.
Collapse
Affiliation(s)
- Noboru Akiyama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Pandolfi A, De Filippis EA. Chronic hyperglicemia and nitric oxide bioavailability play a pivotal role in pro-atherogenic vascular modifications. GENES AND NUTRITION 2007; 2:195-208. [PMID: 18850175 DOI: 10.1007/s12263-007-0050-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2006] [Accepted: 11/10/2006] [Indexed: 02/07/2023]
Abstract
Diabetes is associated with accelerated atherosclerosis and macrovascular complications are a major cause of morbidity and mortality in this disease. Although our understanding of vascular pathology has lately greatly improved, the mechanism(s) underlying enhanced atherosclerosis in diabetes remain unclear. Endothelial cell dysfunction is emerging as a key component in the pathophysiology of cardiovascular abnormalities associated with diabetes. Although it has been established that endothelium plays a critical role in overall homeostasis of the vessels, vascular smooth muscle cells (vSMC) in the arterial intima have a relevant part in the development of atherosclerosis in diabetes. However, high glucose induced alterations in vSMC behaviour are not fully characterized. Several studies have reported that impaired nitric oxide (NO) synthesis and/or actions are often present in diabetes and endothelial dysfunction. Furthermore, although endothelial cells are by far the main site of vascular NO synthesis, vSMC do express nitric oxyde synthases (NOSs) and NO synthesis in vSMC might be important in vessel's function. Although it is known that vSMC contribute to vascular pathology in diabetes by their change from a quiescent state to an activated proliferative and migratory phenotype (termed phenotypic modulation), whether this altered phenotypic modulation might also involve alterations in the nitrergic systems is still controversial. Our recent data indicate that, in vivo, chronic hyperglycemia might induce an increased number of vSMC proliferative clones which persist in culture and are associated with increased eNOS expression and activity. However, upregulation of eNOS and increased NO synthesis occur in the presence of a marked concomitant increase of O(2-) production. Since NO bioavailabilty might not be increased in high glucose stimulated vSMC, it is tempting to hypothesize that the proliferative phenotype observed in cells from diabetic rats is associated with a redox imbalance responsible quenching and/or trapping of NO, with the consequent loss of its biological activity. This might provide new insight on the mechanisms responsible for accelerated atherosclerosis in diabetes.
Collapse
Affiliation(s)
- Assunta Pandolfi
- Aging Research Center, Ce.S.I., "Gabriele D'Annunzio" University Foundation, Department of Biomedical Science, University of "G. D'Annunzio", Room 458, Via Colle dell'Ara, 66013, Chieti-Pescara, Italy,
| | | |
Collapse
|
12
|
Muto A, Fitzgerald TN, Pimiento JM, Maloney S, Teso D, Paszkowiak JJ, Westvik TS, Kudo FA, Nishibe T, Dardik A. Smooth muscle cell signal transduction: implications of vascular biology for vascular surgeons. J Vasc Surg 2007; 45 Suppl A:A15-24. [PMID: 17544020 PMCID: PMC1939976 DOI: 10.1016/j.jvs.2007.02.061] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2007] [Accepted: 02/17/2007] [Indexed: 12/31/2022]
Abstract
Vascular smooth muscle cells exhibit varied responses after vessel injury and surgical interventions, including phenotypic switching, migration, proliferation, protein synthesis, and apoptosis. Although the source of the smooth muscle cells that accumulate in the vascular wall is controversial, possibly reflecting migration from the adventitia, from the circulating blood, or in situ differentiation, the intracellular signal transduction pathways that control these processes are being defined. Some of these pathways include the Ras-mitogen-activated protein kinase, phosphatidylinositol 3-kinase-Akt, Rho, death receptor-caspase, and nitric oxide pathways. Signal transduction pathways provide amplification, redundancy, and control points within the cell and culminate in biologic responses. We review some of the signaling pathways activated within smooth muscle cells that contribute to smooth muscle cell heterogeneity and development of pathology such as restenosis and neointimal hyperplasia.
Collapse
MESH Headings
- Animals
- Apoptosis
- Bone Marrow Cells/metabolism
- Cell Differentiation
- Cell Movement
- Cell Proliferation
- Constriction, Pathologic/metabolism
- Constriction, Pathologic/pathology
- Extracellular Matrix/metabolism
- Humans
- Hyperplasia/metabolism
- Hyperplasia/pathology
- Muscle, Smooth, Vascular/injuries
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Muscle, Smooth, Vascular/surgery
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Protein Kinases/metabolism
- Signal Transduction
- Stem Cells/metabolism
- Vascular Surgical Procedures/adverse effects
Collapse
Affiliation(s)
- Akihito Muto
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
| | - Tamara N Fitzgerald
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
| | - Jose M Pimiento
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
- Saint Mary’s Hospital, Waterbury, CT, USA
| | - Stephen Maloney
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
- Saint Mary’s Hospital, Waterbury, CT, USA
| | - Desarom Teso
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Saint Mary’s Hospital, Waterbury, CT, USA
| | - Jacek J Paszkowiak
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Saint Mary’s Hospital, Waterbury, CT, USA
| | - Tormod S Westvik
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
| | - Fabio A Kudo
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
| | | | - Alan Dardik
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
13
|
Quadros A, Diemer F, Lima T, Abdalla R, Vizotto M, Gottschall CAM, Schaan BD. [Percutaneous coronary intervention in diabetes mellitus: an updated analysis of medical practice]. ARQUIVOS BRASILEIROS DE ENDOCRINOLOGIA E METABOLOGIA 2007; 51:327-33. [PMID: 17505642 DOI: 10.1590/s0004-27302007000200024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2006] [Accepted: 01/04/2007] [Indexed: 05/15/2023]
Abstract
Patients with diabetes and coronary artery disease are frequently considered for myocardial revascularization procedures, aiming at cardiovascular events risk reduction and a better quality of life. In clinical practice, decisions concerning surgery or percutaneous coronary intervention are frequently difficult, because of cases' severity, disease extension and co-morbidities association. Beyond that, the bulk of literature information was generated by subgroup analysis of randomized clinical trials, which were designed for the general population, not for diabetics. The aim of this study was to review literature on coronary percutaneous intervention in diabetic patients, and also to show recent data from the experience in this procedure at the Catheterization Laboratory of the Cardiology Institute of RS.
Collapse
Affiliation(s)
- Alexandre Quadros
- Laboratório de Hemodinâmica, Instituto de Cardiologia do Rio Grande do Sul, Fundação Universitária de Cardiologia, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | |
Collapse
|
14
|
Srivastava S, Ramana KV, Tammali R, Srivastava SK, Bhatnagar A. Contribution of aldose reductase to diabetic hyperproliferation of vascular smooth muscle cells. Diabetes 2006; 55:901-10. [PMID: 16567509 PMCID: PMC3463958 DOI: 10.2337/diabetes.55.04.06.db05-0932] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The objective of this study was to determine whether the polyol pathway enzyme aldose reductase mediates diabetes abnormalities in vascular smooth muscle cell (SMC) growth. Aldose reductase inhibitors (tolrestat or sorbinil) or antisense aldose reductase mRNA prevented hyperproliferation of cultured rat aortic SMCs induced by high glucose. Cell cycle progression in the presence of high glucose was blocked by tolrestat, which induced a G0-G1 phase growth arrest. In situ, diabetes increased SMC growth and intimal hyperplasia in balloon-injured carotid arteries of streptozotocin-treated rats, when examined 7 or 14 days after injury. Treatment with tolrestat (15 mg x kg(-1) x day(-1)) diminished intimal hyperplasia and decreased SMC content of the lesion by 25%. Although tolrestat treatment increased immunoreactivity of the lesion with antibodies raised against protein adducts of the lipid peroxidation product 4-hydroxy trans-2-nonenal, no compensatory increase in lesion fibrosis was observed. Collectively, these results suggest that inhibition of aldose reductase prevents glucose-induced stimulation of SMC growth in culture and in situ. Even though inhibition of aldose reductase increases vascular oxidative stress, this approach may be useful in preventing abnormal SMC growth in vessels of diabetic patients.
Collapse
Affiliation(s)
- Sanjay Srivastava
- Division of Cardiology, Department of Medicine, Delia Baxter Building, 580 S. Preston St., Room 421B, University of Louisville, Louisville, KY 40202, USA.
| | | | | | | | | |
Collapse
|
15
|
Schaan BD, Quadros A, Sarmento-Leite R, Gottschall CAM. Diabetes e implante de stents coronarianos: experiência de um centro de referência em cardiologia intervencionista. ACTA ACUST UNITED AC 2006; 50:38-45. [PMID: 16628273 DOI: 10.1590/s0004-27302006000100006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nosso objetivo foi avaliar a influência do diabetes mellitus (DM) na evolução clínica de pacientes com doença arterial coronariana tratada com implante de stents. Foram implantados 934 stents em 893 pacientes em nossa instituição de 1996 a 2000; 23% destes apresentavam DM. Características clínicas, angiográficas e desfechos clínicos dos pacientes com e sem DM foram prospectivamente incluídos em banco de dados computadorizado. O grupo DM tinha maior idade (61,5 ± 10 vs. 59,8 ± 11 anos; p= 0,04) e prevalência de hipertensão arterial (69% vs. 62%; p= 0,09). A taxa de sucesso clínico do procedimento (estenose residual < 30%, fluxo TIMI 3, ausência de eventos clínicos adversos na internação) foi menor nos pacientes com DM (88% vs. 92%; p= 0,05). Em 1 ano, os pacientes com DM apresentaram maiores taxas de revascularização do vaso-alvo (12,3% vs. 8%; p= 0,06), óbitos (5,4% vs. 2,5%; p= 0,03) e eventos cardiovasculares maiores (ECVM; nova angioplastia, cirurgia, infarto agudo do miocárdio ou óbito: 16,3% vs. 9,3%; p= 0,003). A presença de DM foi independentemente associada a ECVM na análise multivariada (OR: 2,00; IC: 1,253,24; p= 0,004). Concluímos que o DM associa-se a aumento do risco de complicações intra-hospitalares, re-estenose e eventos cardiovasculares maiores em pacientes submetidos ao implante de stents coronarianos.
Collapse
Affiliation(s)
- Beatriz D Schaan
- Laboratório de Hemodinâmica, Instituto de Cardiologia do Rio Grande do Sul, Fundação Universitária de Cardiologia, Porto Alegre, RS.
| | | | | | | |
Collapse
|
16
|
Zimmermann O, Zwaka TP, Marx N, Torzewski M, Bucher A, Guilliard P, Hannekum A, Hombach V, Torzewski J. Serum starvation and growth factor receptor expression in vascular smooth muscle cells. J Vasc Res 2006; 43:157-65. [PMID: 16407661 DOI: 10.1159/000090945] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2005] [Accepted: 10/16/2005] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Smooth muscle cell (SMC) proliferation in atherosclerosis is regulated through the interaction of growth factors like platelet-derived growth factor-BB (PDGF-BB) and insulin-like growth factor-1 (IGF-1) and their receptors (R). We hypothesized that serum starvation of SMCs may affect PDGFbeta-R and IGF-1-R expression and, consequently, the effect of their cognate ligands on SMC survival/proliferation. METHODS AND RESULTS Serum starvation significantly increases PDGFbeta-R but not IGF-1-R mRNA and protein expression in SMCs. PDGF-BB stimulates cell survival but not proliferation in serum-starved SMCs of the synthetic phenotype, whereas SMCs of the contractile phenotype respond to PDGF-BB by a significant increase in proliferation. Immunohistochemical analysis of coronary atherosclerotic lesions reveals PDGFbeta-R expression in SMCs in the lamina fibromuscularis, but not in the media and in healthy parts of the arterial wall. No such differential expression was observed for IGF-1-R. CONCLUSIONS Differential regulation of PDGFbeta-R and IGF-1-R expression by serum starvation might represent a mechanism for the control of SMC survival/proliferation in atherogenesis and restenosis. The distribution of PDGFbeta-Rs and IGF-1-Rs in atherosclerotic lesions may indicate an effect of serum starvation on SMCs in the arterial wall.
Collapse
Affiliation(s)
- Oliver Zimmermann
- Center for Cell and Gene Therapy, and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Tex., USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Seki N, Bujo H, Jiang M, Tanaga K, Takahashi K, Yagui K, Hashimoto N, Schneider WJ, Saito Y. LRP1B is a negative modulator of increased migration activity of intimal smooth muscle cells from rabbit aortic plaques. Biochem Biophys Res Commun 2005; 331:964-70. [PMID: 15882972 DOI: 10.1016/j.bbrc.2005.04.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2005] [Indexed: 10/25/2022]
Abstract
The migration of cultured cultured smooth muscle cells (SMCs) is regulated by the time-specific expression of members of the LDL receptor family (LRs). LRP1B, a member of LRs, modulates the catabolism of PDGF beta-receptor, affecting the migration of SMCs. An involvement of PDGF beta-receptor in atherosclerosis is focused because of its abundant expression in intimal SMCs. Here, in order to know a functional significance of LRP1B in the increased migration of intimal SMCs, the functions of three groups of cultured SMCs with different origins in atherosclerotic arteries were studied. Each group of SMCs (central, marginal or medial SMCs) was isolated from explanted pieces of central or marginal area of thickened intima, or media prepared from rabbit aortic plaques. The LRP1B expression levels were significantly decreased in intimal SMCs, particularly in marginal SMCs, compared to medial SMCs. The expression levels of LRP1B in SMCs were negatively correlated with those of PDGF beta-receptor. The level of PDGF beta-receptor-mediated phosphorylation of ERK 1/2 in central SMCs was increased to 5.2-fold with the functional inhibition of LRP1B using anti-LRP1B IgY. The antibody increased the PDGF-BB-stimulated migration and invasion activities in SMCs. The increase in the PDGF beta-receptor-mediated outgrowth activity of SMCs from the explants was also inhibited by the functional inhibition of LRP1B. These results indicate that LRP1B regulated the migration activity of SMCs through the modulation of PDGF beta-receptor-mediated pathway. The regulation of LRP1B expression is possibly involved in the activated migration of intimal SMCs in the course of atherosclerosis.
Collapse
Affiliation(s)
- Naoto Seki
- Department of Clinical Cell Biology (F5), Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Arampatzis CA, Goedhart D, Serruys PW, Saia F, Lemos PA, de Feyter P. Fluvastatin reduces the impact of diabetes on long-term outcome after coronary intervention--a Lescol Intervention Prevention Study (LIPS) substudy. Am Heart J 2005; 149:329-35. [PMID: 15846273 DOI: 10.1016/j.ahj.2004.03.067] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
BACKGROUND Diabetes increases the risk of developing cardiovascular disease. Patients with diabetes undergoing percutaneous coronary intervention (PCI) show poorer outcomes compared with nondiabetic patients. The aim of this study was to determine the clinical benefit of long-term fluvastatin in patients with diabetes who had undergone a successful PCI. METHODS This subanalysis of a prospective, multicenter, randomized, double-blind, placebo-controlled trial of patients who had undergone PCI and were treated with fluvastatin determined the impact of fluvastatin on the survival-free period of major adverse cardiac events (MACE) (defined as cardiac death, nonfatal myocardial infarction, and reintervention procedure [coronary artery bypass grafting, repeat PCI, PCI for a new lesion]). Patients with baseline total cholesterol levels of 135 to 270 mg/dL (3.5-7.0 mmol/L) and triglyceride levels of 400 mg/dL (4.5 mmol/L) were randomized at discharge either to fluvastatin (n = 844) or to placebo (n = 833); follow-up was 3 to 4 years. Among these patients, there were 202 with diabetes (120 on fluvastatin, 82 placebo) and 1475 without diabetes (724 on fluvastatin, 751 on placebo). The primary clinical outcome was survival time free of MACE and MACE excluding restenosis. RESULTS The presence of diabetes increased the risk of MACE by almost 2-fold in placebo-treated patients (RR 1.78, 95% CI 1.20-2,64, P = .0045). In contrast, in diabetic patients treated with fluvastatin, the risk of MACE was not significantly different from that in patients without diabetes. Fluvastatin reduced the risk of MACE in diabetic patients by 51% (P = .0088). CONCLUSIONS Diabetes is a consistent clinical predictor of cardiovascular complications and fluvastatin reduces the increased incidence of long-term adverse complications associated with the presence of diabetes.
Collapse
|
19
|
Miyashita Y, Koide N, Oyama T, Itoh Y, Shirai K. Apoptosis Induced by 7-Ketocholesterol is Enhanced in Smooth Muscle Cells Derived from OLETF Rats. J Atheroscler Thromb 2005; 12:92-7. [PMID: 15942119 DOI: 10.5551/jat.12.92] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
To clarify whether an increased proliferative potential of vascular smooth muscle cells (SMC) under diabetic conditions augments the susceptibility of the cells to 7-ketocholesterol-induced apoptosis, we investigated the difference in sensitivity to 7-ketocholesterol between SMC obtained from diabetic Otsuka Long-Evans Tokushima fatty (OLETF) rats and the control Long-Evans Tokushima Otsuka (LETO) rats. The outgrowth rate from aortic wall explants and cell proliferation were higher in SMC derived from OLETF rats (OLETF-derived SMC) compared to those from LETO rats (LETO-derived SMC). When 7-ketocholesterol was added to SMC, the amount of fragmented DNA increased significantly in OLETF-derived compared to LETO-derived SMC. The amount of fragmented DNA induced by 7-ketocholesterol decreased significantly in both OLETF- and LETO-derived SMC when they were incubated without fetal bovine serum. By adding PDGF-BB to LETO-derived SMC, the amount of fragmented DNA induced by 7-ketocholesterol increased significantly. These results suggest that apoptosis of SMC induced by 7-ketocholesterol may be accelerated when SMC acquire a high proliferative potential by prolonged exposure to diabetic condition.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/pathology
- Apoptosis/drug effects
- Apoptosis/genetics
- Arteriosclerosis/etiology
- Arteriosclerosis/pathology
- Cell Proliferation/drug effects
- Cells, Cultured
- DNA Fragmentation/drug effects
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/pathology
- Enzyme Inhibitors/pharmacology
- Enzyme-Linked Immunosorbent Assay
- In Vitro Techniques
- Ketocholesterols/pharmacology
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Rats
- Rats, Inbred OLETF
- Rats, Long-Evans
Collapse
Affiliation(s)
- Yoh Miyashita
- Center of Diabetes, Endocrine and Metabolism, Sakura Hospital, Toho University School of Medicine, 564 Shimoshizu, Sakura-shi, Chiba 285-8741, Japan.
| | | | | | | | | |
Collapse
|
20
|
Maca T, Schillinger M, Hamwi A, Mlekusch W, Sabeti S, Wagner O, Minar E. Insulin, C-Peptide, and Restenosis after Femoral Artery Balloon Angioplasty in Type II Diabetic and Nondiabetic Patients. J Vasc Interv Radiol 2005; 16:31-5. [PMID: 15640407 DOI: 10.1097/01.rvi.0000136030.26074.33] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
PURPOSE Endogenous and exogenous insulin is suggested to stimulate hypertrophic wound-healing responses and therefore may promote neointimal hyperplasia and restenosis after balloon angioplasty. The ratio of C-peptide to insulin reflects endogenous insulin secretion. In diabetic patients with insulin substitution, lower ratios display a higher proportion of exogenous insulin. The association and interaction of insulin and C-peptide with restenosis after percutaneous transluminal angioplasty (PTA) was investigated in type II diabetic and nondiabetic patients. MATERIALS AND METHODS The study group included 76 patients (median age, 68 years; interquartile range [IQR], 58-74 years; 55 men [72%]; 31 patients [41%] with type II diabetes) with intermittent claudication (n = 49; 64%) or critical limb ischemia (n = 27; 36%) who underwent primary successful femoral PTA. C-peptide and insulin levels were measured at baseline, and patients were followed to determine restenosis (> or =50%) at 12 months by color-coded duplex sonography. RESULTS Restenosis was found in 34 patients (45%) at 12 months. Patients with restenosis had higher insulin levels (median, 21.3 microU/mL IQR, 11.3-35.5 microU/mL) and a lower C-peptide/insulin ratio (median, 16; IQR, 10-21) compared with patients without restenosis (median insulin level, 11.6 microU/mL; IQR, 9.1-22.0 microU/mL [P = .008]; median ratio, 19 [IQR, 17-25], P = .039). In nondiabetic patients, insulin levels were significantly associated with restenosis (P = .046), whereas the ratio of C-peptide to insulin showed no association with restenosis. In patients with type II diabetes (n = 31; 41%), in contrast, the C-peptide/insulin ratio was associated with restenosis (P = .047), whereas insulin levels showed no significant association with restenosis (P = .14). CONCLUSIONS Insulin levels and the C-peptide/insulin ratio were associated with restenosis after femoral PTA. Exogenous and endogenous insulin may play a role in the pathogenesis of recurrent lumen loss after balloon angioplasty.
Collapse
Affiliation(s)
- Thomas Maca
- Department of Internal Medicine II, Division of Angiology, University of Vienna Medical School, Waehringer Guertel 18-20, A-1090 Vienna, Austria.
| | | | | | | | | | | | | |
Collapse
|
21
|
Osman A, Otero J, Brizolara A, Waxman S, Stouffer G, Fitzgerald P, Uretsky BF. Effect of rosiglitazone on restenosis after coronary stenting in patients with type 2 diabetes. Am Heart J 2004; 147:e23. [PMID: 15131558 DOI: 10.1016/j.ahj.2003.12.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Thiazolidinediones have been shown to have an antiproliferative vascular effect in experimental models. We sought to study the effect of rosiglitazone on in-stent restenosis in patients with established type 2 diabetes. METHODS Patients with treated type 2 diabetes (mean duration 5.5 +/- 7.5 years) referred for coronary stenting were randomized in a double-blind fashion to receive oral rosiglitazone or placebo for 6 months. Quantitative coronary angiography and intravascular ultrasound data were obtained at baseline and follow-up. Plasma plasminogen activator inhibitor-1 levels were prospectively measured. RESULTS Sixteen patients were enrolled. There were no significant differences in follow-up in-stent luminal diameter stenosis measured by quantitative coronary angiography or in-stent luminal area stenosis and neointimal volume index obtained by intravascular ultrasound, nor were there any differences in plasma plasminogen activator inhibitor-1 levels after long-term use despite improvement in diabetes control and insulin sensitivity. CONCLUSIONS Rosiglitazone, given at the time of stent implantation in treated diabetics, did not reduce in-stent restenosis in this small series. The vascular biological effects of this agent await further clarification in humans and evaluation in larger clinical trials.
Collapse
Affiliation(s)
- Abdulfatah Osman
- Division of Cardiology, University of Texas Medical Branch, Galveston, Texas, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Lemström KB, Nykänen AI, Tikkanen JM, Krebs R, Sihvola RK, Kallio EA, Olivier R, Koskinen PK. Role of angiogenic growth factors in transplant coronary artery disease. Ann Med 2004; 36:184-93. [PMID: 15181974 DOI: 10.1080/07853890310025243] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Transplant coronary artery disease (TxCAD) as a manifestation of chronic rejection is a major limitation to long-term survival of heart transplant recipients. Although the exact molecular and cellular mechanisms contributing to neointimal formation are unknown, it has been generally believed that smooth muscle cells (SMC) of donor origin migrate from the media into the subendothelial layer of the vascular wall, where SMC proliferate and synthesize extracellular matrix resulting in intimal thickening. However, recent observations indicate that hematopoietic and vascular progenitor cells derived from recipient bone marrow may contribute to the arteriosclerotic lesion formation in the coronary arteries of the transplant. On the other hand, studies on postnatal hematopoiesis indicate that angiogenic growth factors such as vascular endothelial growth factor (VEGF) and angiopoietin-1 (Ang1) may regulate the recruitment of these cells into distant organs. Furthermore, embryonic VEGFR-2 /CD34+ stem cells may serve as vascular progenitor cells and their differentiation into endothelial cells and SMC may be regulated by VEGF and platelet-derived growth factor (PDGF), respectively. In this review, we discuss the role of angiogenic growth factors such as VEGF, Ang, and PDGF in the recruitment of hematopoietic and vascular progenitor cells in TxCAD and suggest novel therapies targeted at homing, differentiation and proliferation of these cells in the allograft.
Collapse
Affiliation(s)
- Karl B Lemström
- Cardiopulmonary Research Group, Transplantation Laboratory, University of Helsinki, and Helsinki University Central Hospital, Helsinki, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Pandolfi A, Grilli A, Cilli C, Patruno A, Giaccari A, Di Silvestre S, De Lutiis MA, Pellegrini G, Capani F, Consoli A, Felaco M. Phenotype modulation in cultures of vascular smooth muscle cells from diabetic rats: association with increased nitric oxide synthase expression and superoxide anion generation. J Cell Physiol 2003; 196:378-85. [PMID: 12811832 DOI: 10.1002/jcp.10325] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Proliferative modification of vascular smooth muscle cell (vSMC) and impaired bioavailability of nitric oxide (NO) have both been proposed among the mechanisms linking diabetes and atherosclerosis. However, diabetes induced modifications in phenotype and nitric oxide synthase(s) (NOS) expression and activity in vSMC have not been fully characterized. In this study, cell morphology, proliferative response to serum, alpha-SMactin levels, eNOS expression and activity, cGMP intracellular content, and superoxide anion release were measured in cultures of vSMC obtained from aorta medial layer of ten diabetic (90% pancreatectomy, DR) and ten control (sham surgery, CR) rats. Vascular SMC from DR showed a less evident "hill and valley" culture morphology, increased growth response to serum, greater saturation density, and lower levels of alpha-SMactin. In the same cells, as compared to CR cells, eNOS mRNA levels and NOS activity were increased, while intracellular cGMP level was lower and superoxide anion production was significantly greater. These data indicate that chronic hyperglycemia might induce, in the vascular wall, an increased number of vSMC proliferative clones which persist in culture and are associated with increased eNOS expression and activity. However, upregulation of eNOS and increased NO synthesis occur in the presence of a marked concomitant increase of O(2-) production. Since NO bioavailability, as reflected by cGMP levels, was not increased in DR cells, it is tempting to hypothesize that the proliferative phenotype observed in DR cells is associated with a redox imbalance responsible quenching and/or trapping of NO, with the consequent loss of its biological activity.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic
- Blood Glucose/analysis
- Blood Physiological Phenomena
- Cell Count
- Cells, Cultured
- Citrulline/biosynthesis
- Cytoskeleton/ultrastructure
- Diabetes Mellitus/etiology
- Diabetes Mellitus/genetics
- Diabetes Mellitus/metabolism
- Diabetes Mellitus/pathology
- Diabetes Mellitus/physiopathology
- Fatty Acids, Nonesterified/blood
- Flow Cytometry
- Insulin/blood
- Male
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/physiology
- Nitric Oxide Synthase/genetics
- Nitric Oxide Synthase/metabolism
- Nitric Oxide Synthase Type III
- Pancreatectomy/methods
- Phenotype
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Superoxides/metabolism
Collapse
|
24
|
Kornowski R, Mintz GS, Abizaid A, Leon MB. Intravascular ultrasound observations of atherosclerotic lesion formation and restenosis in patients with diabetes mellitus. INTERNATIONAL JOURNAL OF CARDIOVASCULAR INTERVENTIONS 2003; 2:13-20. [PMID: 12623382 DOI: 10.1080/acc.2.1.13.20] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Coronary artery disease is more aggressive in diabetic patients than in nondiabetics; they have more diffuse disease, higher mortality rates and worse clinical outcomes after coronary interventions. Intravascular ultrasound (IVUS) produces transmural tomographic images of the coronary arteries in vivo. Recent IVUS studies have provided new insights into the mechanisms of stenosis formation and restenosis in both nondiabetic and diabetic patients. Arterial remodeling is defined as a change in arterial area. During atherogenesis, an increase in arterial area usually accompanies plaque accumulation to delay lumen compromise. Stenosis formation is related to: (a) the rate of plaque accumulation versus the rate of positive remodeling; and (b) the limits and ultimate failure of positive remodeling. However, there is a marked variability in remodeling. IVUS studies have suggested that remodeling may be impaired in some diabetic patients during atherogenesis. Following non-stent catheter-based interventions, serial (post-intervention and follow-up) IVUS studies have shown that the change in lumen area correlates better with the change in arterial area (remodeling) than with the change in plaque area (neointimal hyperplasia). In some patients, a positive remodeling response mitigates against the increase in plaque area to limit late lumen loss and restenosis. Neointimal hyperplasia is exaggerated in diabetic patients. Despite this, there is a reduced frequency of positive remodeling, potentially similar to the impaired positive remodeling in some diabetic patients during atherogenesis. Failed or inadequate arterial remodeling may contribute to the pathogenesis and natural history of atherosclerotic coronary artery disease in diabetic patients.
Collapse
Affiliation(s)
- Ran Kornowski
- The Cardiac Catheterization and the, Intravascular Ultrasound Imaging Laboratories, Washington Hospital Center, Washington DC, USA
| | | | | | | |
Collapse
|
25
|
Kanzaki T, Otabe M. Latent transforming growth factor-beta binding protein-1, a component of latent transforming growth factor-beta complex, accelerates the migration of aortic smooth muscle cells in diabetic rats through integrin-beta3. Diabetes 2003; 52:824-8. [PMID: 12606526 DOI: 10.2337/diabetes.52.3.824] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Aortic smooth muscle cells (SMCs) of diabetic animals have unique properties, including the overexpression of transforming growth factor-beta (TGF-beta) type II receptor, fibronectin, and platelet-derived growth factor beta-receptor. TGF-beta1 is produced and secreted as latent high-molecular weight complex consisting of mature TGF-beta1, latency-associated peptide (LAP), and a latent TGF-beta1 binding protein (LTBP-1). LAP has an important function in the latency of TGF-beta complex, but the role of LTBP-1 is not known in diabetic angiopathy. SMC migration from the medial layer to the intimal layer of an artery is an initial major process of the formation of intimal thickening of an artery. Migration activities of SMCs from diabetic rat with 1-500 pg/ml of LTBP-1 increased significantly compared with that without LTBP-1. LTBP-1 at 10-500 pg/ml stimulated the migration of diabetic SMCs more than SMCs from control rat. An anti-integrin-beta(3) antibody reduced LTBP-1-stimulated migration of diabetic SMCs to 51% compared with no antibody, but it did not reduce that of control SMCs. Furthermore, cross-linking experiments show that LTBP-1 binds integrin-beta(3) in diabetic SMCs much more than in control SMCs in coincidence with the increase of integrin-beta(3) in diabetic aorta by immunohistochemistry. Taken together, these observations suggest that LTBP-1 plays a critical role in intimal thickening of diabetic artery through the acceleration of SMC migration via integrin-beta(3).
Collapse
Affiliation(s)
- Tetsuto Kanzaki
- Department of Internal Medicine, Kohnodai Hospital, National Center of Neurology and Psychiatry, 1-7-1 Kohnodai, Ichikawa City 272-8516, Japan.
| | | |
Collapse
|
26
|
Goto M, Yamauchi Y, Kurosaki E, Azuma H. Possible involvement of facilitated polyol pathway in augmentation of intimal hyperplasia in rabbits with alloxan-induced hyperglycemia. J Cardiovasc Pharmacol 2003; 41:265-75. [PMID: 12548088 DOI: 10.1097/00005344-200302000-00016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Present experiments were designed to investigate whether the facilitated polyol pathway is involved in the augmentation of intimal hyperplasia with hyperglycemia. Twelve weeks after a single bolus intravenous injection of alloxan (100 mg/kg) or saline, rabbits underwent a unilateral endothelial denudation of the carotid artery. Intimal hyperplasia was evident 4 weeks after denudation and significantly augmented in hyperglycemic animals treated with alloxan. This effect was accompanied by the enhanced accumulation of endogenous NOS inhibitors (N(G)-monomethyl-l-arginine [l-NMMA] and asymmetric, N(G),N(G)-dimethyl-l-arginine [ADMA]) in regenerated endothelial cells, impairment of NO production and release, and enhanced accumulation of endothelin-1 (ET-1) within the vessel wall. Sorbitol levels in aortic endothelial cells and within the smooth muscle layer were significantly increased with hyperglycemia. All these changes associated with hyperglycemia were significantly reduced in animals treated with the selective aldose reductase inhibitor fidarestat (3 mg/kg/d). These findings suggest that the facilitated polyol pathway possibly plays an important role for the augmentation of intimal hyperplasia caused by the hyperglycemic state.
Collapse
Affiliation(s)
- Moritaka Goto
- Department of Molecular Design, Institute of Biomaterials and Bioengineering, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | |
Collapse
|
27
|
Wang W, Campos AH, Prince CZ, Mou Y, Pollman MJ. Coordinate Notch3-hairy-related transcription factor pathway regulation in response to arterial injury. Mediator role of platelet-derived growth factor and ERK. J Biol Chem 2002; 277:23165-71. [PMID: 11971902 DOI: 10.1074/jbc.m201409200] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The Notch family of receptors and downstream effectors plays a critical role in cell fate determination during vascular ontogeny. Moreover, the human cerebral autosomal dominant artriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) syndrome of premature stroke and dementia is a heritable arteriopathy with alterations in vascular smooth muscle cells (VSMCs) resulting from mutations within Notch3. However, the expression and regulation of the Notch and hairy-related transcription factor (HRT) pathway in adult VSMCs in vitro and in vivo remain poorly characterized. The present study documents that the well-described modulation of VSMC fate in response to vascular injury and growth factor activation involves a coordinate regulation of the Notch and HRT pathways. Furthermore, platelet-derived growth factor promotes a similar coordinate down-regulation of the Notch receptors and HRT genes in cultured VSMCs via an ERK-dependent signaling pathway. Moreover, we established that HRT1 and HRT2 are direct downstream target genes of Notch3 signaling in VSMCs and determined that the activity of the nuclear protein RBP-Jk is essential for their regulation. These findings provide initial insight into the context- and cell type-dependent coordinate regulation of Notch3 and downstream HRT transcriptional pathway effector genes in VSMCs in vitro and in vivo that may have important implications for understanding the role of Notch signaling in human health and vascular disease.
Collapse
Affiliation(s)
- Wenli Wang
- Cardiovascular Research Institute, Morehouse School of Medicine, Research Wing, Atlanta, Georgia 30310, USA
| | | | | | | | | |
Collapse
|
28
|
Park SH, Marso SP, Zhou Z, Foroudi F, Topol EJ, Lincoff AM. Neointimal hyperplasia after arterial injury is increased in a rat model of non-insulin-dependent diabetes mellitus. Circulation 2001; 104:815-9. [PMID: 11502708 DOI: 10.1161/hc3301.092789] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The key biological determinants that promote restenosis in the setting of diabetes have not been elucidated. There is no accepted animal model to study restenosis in diabetes. METHODS AND RESULTS We evaluated 2 models of diabetes mellitus: (1) streptozotocin (STZ)-treated Sprague-Dawley rats (type I diabetes) versus regular Sprague-Dawley rats and (2) obese Zucker rats (type II diabetes) versus lean Zucker rats. Neointimal hyperplasia was assessed after carotid balloon injury at 21 days by computerized morphometry. There was no difference in neointimal area in the STZ-treated rats compared with controls, irrespective of insulin administration or dose of STZ. Neointimal area was increased >2-fold in obese Zucker rats compared with lean Zucker rats (0.21+/-0.06 versus 0.08+/-0.03 mm(2), P<0.01). The neointimal area was markedly increased in the obese Zucker rats 7 days after injury (0.058+/-0.024 versus 0.033+/-0.009 mm(2), P<0.05) and persisted through 21 days. In both obese and lean Zucker rats, cell proliferation peaked in the media at 3 days (118.66+/-84.28 versus 27.50+/-12.75 bromodeoxyuridine-labeled cells per cross section). In the intima, cell proliferation markedly increased beginning at day 3 and persisted through day 14 in the obese and lean Zucker rats (202.27+/-98.86 versus 35.71+/-20.54 bromodeoxyuridine-labeled cells at 7 days). CONCLUSIONS The type II diabetic rat model, typifying insulin resistance, is associated with a propensity for neointima. The obese Zucker rat model may be an ideal diabetic model to further characterize the diabetic vascular response to injury.
Collapse
Affiliation(s)
- S H Park
- Department of Cardiology, The Cleveland Clinic Foundation, Cleveland, Ohio 44195-5066, USA
| | | | | | | | | | | |
Collapse
|
29
|
Ostman A, Heldin CH. Involvement of platelet-derived growth factor in disease: development of specific antagonists. Adv Cancer Res 2001; 80:1-38. [PMID: 11034538 DOI: 10.1016/s0065-230x(01)80010-5] [Citation(s) in RCA: 151] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Platelet-derived growth factor (PDGF) is a family of dimeric isoforms that stimulates, e.g., growth, chemotaxis and cell shape changes of various connective tissue cell types and certain other cells. The cellular effects of PDGF isoforms are exerted through binding to two structurally related tyrosine kinase receptors. Ligand binding induces receptor dimerization and autophosphorylation. This enables a number of SH2 domain containing signal transduction molecules to bind to the receptors, thereby initiating various signaling pathways. PDGF isoforms have important roles during the embryonic development, particularly in the formation of connective tissue in various organs. In the adult, PDGF stimulates wound healing. Overactivity of PDGF has been implicated in certain disorders, including fibrotic conditions, atherosclerosis, and malignancies. Different kinds of PDGF antagonists are currently being developed and evaluated in different animal disease models, as well as in clinical trials.
Collapse
Affiliation(s)
- A Ostman
- Ludwig Institute for Cancer Research, Biomedical Center, Uppsala, Sweden
| | | |
Collapse
|
30
|
Yasuda Y, Nakamura J, Hamada Y, Nakayama M, Chaya S, Naruse K, Nakashima E, Kato K, Kamiya H, Hotta N. Role of PKC and TGF-beta receptor in glucose-induced proliferation of smooth muscle cells. Biochem Biophys Res Commun 2001; 281:71-7. [PMID: 11178962 DOI: 10.1006/bbrc.2001.4310] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The role of protein kinase C (PKC) and transforming growth factor (TGF)-beta in the proliferation of vascular smooth muscle cells (SMCs) under a high glucose condition was investigated. [3H]-thymidine incorporation under 20 mM glucose was significantly accelerated compared with that under 5.5 mM glucose, and this increase was inhibited by an anti-TGF-beta antibody or a PKC-beta specific inhibitor, LY333531. The amount of active and total TGF-beta1 in the conditioned media did not differ between 5.5 and 20 mM glucose. However, the expression of TGF-beta receptor type II under 20 mM glucose was significantly increased, but that of the TGF-beta receptor type I was not. This increased expression of the TGF-beta receptor type II was prevented by LY333531. These observations suggest that the increased expression of the TGF-beta receptor type II via PKC-beta plays an important role in the accelerated proliferation of SMCs under a high glucose condition, leading to the development of diabetic macroangiopathy.
Collapse
MESH Headings
- Activin Receptors, Type I
- Animals
- Aorta/metabolism
- Cell Division
- Cells, Cultured
- Culture Media, Conditioned/metabolism
- DNA/biosynthesis
- Dose-Response Relationship, Drug
- Enzyme Inhibitors/pharmacology
- Glucose/metabolism
- Glucose/pharmacology
- Immunoblotting
- Indoles/pharmacology
- Maleimides/pharmacology
- Muscle, Smooth/cytology
- Muscle, Smooth/metabolism
- Protein Kinase C/physiology
- Protein Serine-Threonine Kinases/metabolism
- RNA, Messenger/metabolism
- Rats
- Receptor, Transforming Growth Factor-beta Type I
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Transforming Growth Factor beta/metabolism
- Receptors, Transforming Growth Factor beta/physiology
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Y Yasuda
- Third Department of Internal Medicine, Nagoya University School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Hirata A, Igarashi M, Yamaguchi H, Suwabe A, Daimon M, Kato T, Tominaga M. Nifedipine suppresses neointimal thickening by its inhibitory effect on vascular smooth muscle cell growth via a MEK-ERK pathway coupling with Pyk2. Br J Pharmacol 2000; 131:1521-30. [PMID: 11139427 PMCID: PMC1572490 DOI: 10.1038/sj.bjp.0703730] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The aim of this study was to determine whether nifedipine could suppress an atherogenic process such as balloon-injured intimal thickening in vivo and the proliferation of vascular smooth muscle cells (VSMC) in vitro. First, we examined the in vivo effect of nifedipine to determine whether it could suppress intimal thickening induced by balloon catheterization. Sprague-Dawley (SD) rats were divided into three groups (L, nifedipine 0.3 mg kg(-1) day(-1); H, nifedipine 3 mg kg(-1) day(-1); C, no nifedipine), and Alzet((R)) osmotic pumps were implanted in their backs for continuous administration. The neointimal layers were completely occupied by proliferated VSMC, and the area ratios of neointima/media treated with nifedipine significantly decreased dose-dependently compared to those of the control. Neither blood pressure nor lipid levels changed among the three groups. We next evaluated the in vitro effect of nifedipine on the proliferation of cultured rat VSMC. Nifedipine decreased the values of [(3)H]-thymidine incorporation and total cellular protein content as well as the levels of phosphorylated extracellular signal-regulated protein kinase (ERK) 1/2, mitogen-activated protein kinase kinase (MEK) 1/2, and even the phosphorylation of Pyk2, in dose-dependent fashions. In addition, nifedipine suppressed the levels of proliferative cell nuclear antigen (PCNA) dose-dependently in both VSMC and balloon-injured thoracic aortae. These results indicate that nifedipine has an inhibitory effect on intimal thickening by attenuating intimal VSMC proliferation, suggesting that nifedipine could be effective for preventing the progression of atherosclerotic plaque as in restenosis after angioplasty.
Collapse
MESH Headings
- Actins/analysis
- Animals
- Aorta, Thoracic/chemistry
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/pathology
- Blood Pressure/drug effects
- Body Weight/drug effects
- Calcium Channel Blockers/pharmacology
- Carotid Arteries/drug effects
- Carotid Arteries/pathology
- Catheterization
- Cell Division/drug effects
- Cell Survival/drug effects
- Cells, Cultured
- Dose-Response Relationship, Drug
- Focal Adhesion Kinase 2
- Heart Rate/drug effects
- Immunohistochemistry
- Lipids/blood
- MAP Kinase Signaling System/drug effects
- Male
- Mitogen-Activated Protein Kinases/drug effects
- Mitogen-Activated Protein Kinases/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Nifedipine/pharmacology
- Phosphorylation/drug effects
- Proliferating Cell Nuclear Antigen/drug effects
- Proliferating Cell Nuclear Antigen/metabolism
- Protein Serine-Threonine Kinases/drug effects
- Protein Serine-Threonine Kinases/metabolism
- Protein-Tyrosine Kinases/drug effects
- Protein-Tyrosine Kinases/metabolism
- Rats
- Rats, Sprague-Dawley
- Tunica Intima/chemistry
- Tunica Intima/drug effects
- Tunica Intima/pathology
- Tunica Media/chemistry
- Tunica Media/drug effects
- Tunica Media/pathology
Collapse
Affiliation(s)
- Akihiko Hirata
- Department of Laboratory Medicine, Yamagata University School of Medicine, 2-2-2, Iida-nishi, Yamagata 990-9585, Japan
| | - Masahiko Igarashi
- Third Department of Internal Medicine, Yamagata University School of Medicine, 2-2-2, Iida-nishi, Yamagata 990-9585, Japan
- Author for correspondence:
| | - Hiroshi Yamaguchi
- Third Department of Internal Medicine, Yamagata University School of Medicine, 2-2-2, Iida-nishi, Yamagata 990-9585, Japan
| | - Akira Suwabe
- Department of Laboratory Medicine, Yamagata University School of Medicine, 2-2-2, Iida-nishi, Yamagata 990-9585, Japan
| | - Makoto Daimon
- Third Department of Internal Medicine, Yamagata University School of Medicine, 2-2-2, Iida-nishi, Yamagata 990-9585, Japan
| | - Takeo Kato
- Third Department of Internal Medicine, Yamagata University School of Medicine, 2-2-2, Iida-nishi, Yamagata 990-9585, Japan
| | - Makoto Tominaga
- Department of Laboratory Medicine, Yamagata University School of Medicine, 2-2-2, Iida-nishi, Yamagata 990-9585, Japan
| |
Collapse
|
32
|
Takagi T, Akasaka T, Yamamuro A, Honda Y, Hozumi T, Morioka S, Yoshida K. Troglitazone reduces neointimal tissue proliferation after coronary stent implantation in patients with non-insulin dependent diabetes mellitus: a serial intravascular ultrasound study. J Am Coll Cardiol 2000; 36:1529-35. [PMID: 11079654 DOI: 10.1016/s0735-1097(00)00895-0] [Citation(s) in RCA: 147] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES The aim of the present study was to determine whether troglitazone reduces neointimal tissue proliferation after coronary stent implantation in patients with non-insulin dependent diabetes mellitus (NIDDM). BACKGROUND Increased in-stent restenosis in patients with diabetes mellitus is due to accelerated neointimal tissue proliferation after coronary stent implantation. Troglitazone inhibits intimal hyperplasia in experimental animal models. METHODS We studied 62 stented lesions in 52 patients with plasma glucose levels (PG) > or = 11.1 mmol/liter at 2 h after 75 g oral glucose load. The study patients were randomized into two groups: the troglitazone group of 25 patients with 29 stents, who were treated with 400 mg of troglitazone, and the control group of 27 patients with 33 stents. All patients underwent oral glucose tolerance tests before and after their six-month treatment period. The sum of PG (sum of PG) and the sum of insulin levels (sum of IRI) were measured. Serial (postintervention and at six-month follow-up) intravascular ultrasound studies were performed. Cross-sectional images within stents were taken at every 1 mm, using an automatic pullback. Stent areas (SA), lumen areas (LA), and intimal areas (IA = SA - LA) were measured and averaged over a number of selected image slices. The intimal index was calculated as intimal index = averaged IA/averaged SA x 100%. RESULTS There were no differences between the two groups before treatment in sum of PG (31.35 +/- 3.07 mmol/liter vs. 32.89 +/- 4.87 mmol/liter, respectively, p = 0.2998) and sum of IRI (219.6 +/- 106.2 mU/liter vs. 209.2 +/- 91.6 mU/liter, respectively, p = 0.8934). However, reductions in sum of PG at the six-month follow-up in the troglitazone group were significantly greater than those in the control group (-21.4 +/- 8.8% vs. -4.5 +/- 7.4%, respectively, p < 0.0001). Likewise, decreases in sum of IRI were greater in the troglitazone-treated group (-31.4 +/- 17.9% vs. -1.9 +/- 15.1%, respectively, p < 0.0001). Although, there were no differences between the two groups in SA at postintervention (7.4 +/- 2.2 mm2 vs. 7.3 +/- 1.7 mm2, respectively, p = 0.9482) and at follow-up (7.3 +/- 2.3 mm2 vs. 7.3 +/- 1.8 mm2, respectively, p = 0.2307), the LA at follow-up in the troglitazone group was significantly greater than that in the control group (5.3 +/- 1.7 mm2 vs. 3.7 +/- 1.7 mm2, respectively, p = 0.0002). The IA at follow-up in the troglitazone group was significantly smaller than that in the control group (2.0 +/- 0.9 mm2 vs. 3.5 +/- 1.8 mm2, respectively, p < 0.0001). This was also true for intimal index (27.1 +/- 11.5% vs. 49.0 +/- 14.4%, respectively, p < 0.0001). CONCLUSIONS Serial intravascular ultrasound assessment shows that administration of troglitazone reduces neointimal tissue proliferation after coronary stent implantation in patients with NIDDM.
Collapse
Affiliation(s)
- T Takagi
- Division of Cardiology, Kobe General Hospital, Japan.
| | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Diabetes mellitus has a negative impact on mortality and morbidity following catheter-based coronary procedures as well as coronary artery bypass surgery. Increased restenosis remains the main limitation of catheter-based coronary intervention among diabetes mellitus in addition to accelerated atherosclerosis lesion progression in other untreated coronary sites. Determinants such as excess restenosis, high atherosclerosis burden, lesion complexity, small target vessel size, and accelerated coronary atherosclerosis in remote sites may favor the surgical strategy in most cases of diabetic multivessel disease. The importance of periprocedural adjunctive pharmacotherapy, specifically with the use of antiplatelet and long-term antilipidemic treatment, was shown to improve outcomes in diabetics undergoing percutaneous coronary interventions. The purpose of the review is to examine potential mechanisms causing more restenosis in diabetics, the clinical outcomes of patients with diabetes after coronary interventions including stenting, the treatment alternatives of diabetic patients with diffuse coronary artery disease, including coronary bypass surgery, and current understanding of the benefit of adjunctive pharmacology on clinical outcomes after coronary interventions among diabetics.
Collapse
Affiliation(s)
- R Kornowski
- Cardiac Catheterization Laboratory and Cardiovascular Research Foundation, Washington Hospital Center, Washington, DC 20010, USA.
| | | |
Collapse
|
34
|
Tamura K, Kanzaki T, Tashiro J, Yokote K, Mori S, Ueda S, Saito Y, Morisaki N. Increased atherogenesis in Otsuka Long-Evans Tokushima fatty rats before the onset of diabetes mellitus: association with overexpression of PDGF beta-receptors in aortic smooth muscle cells. Atherosclerosis 2000; 149:351-8. [PMID: 10729385 DOI: 10.1016/s0021-9150(99)00349-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
The mechanism of diabetic macroangiopathy was studied from the view point of phenotypic change of arterial smooth muscle cells (SMC). Otsuka Long-Evans Tokushima fatty (OLETF) rat, an animal model of non-insulin dependent diabetes mellitus (NIDDM), develops spontaneous persistent hyperglycemia after the age of 18 weeks. Medial SMC in OLETF rats expressed more platelet-derived growth factor (PDGF) beta-receptor and fibronectin at the protein level than those from control, Long-Evans Tokushima Otsuka (LETO) rats, not only after but also before the onset of diabetes mellitus. Cultured SMC from OLETF rats more strongly responded specifically to the mitogenic stimuli of PDGF-AB and PDGF-BB and also expressed PDGF beta-receptor more intensely compared with those from LETO rats. PDGF is known to be the main contributor to the intimal thickening induced by balloon catheter injury, which is one of several forms of arterial injuries. Intimal thickening of carotid arteries in OLETF rats after balloon catheter injury increased compared with that in LETO rats before the onset of diabetes mellitus. In in vitro culture system, fibronectin synthesis was stimulated by transforming growth factor-beta1(TGF-beta1) in SMC from OLETF rats, but not in those from LETO rats, suggesting that SMC from OLETF rats respond to TGF-beta1. These results indicate that overexpression of PDGF beta-receptor and fibronectin in medial SMC plays an important role in the accelerated intimal thickening before the onset of diabetes mellitus in OLETF rats.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/cytology
- Aorta, Thoracic/pathology
- Becaplermin
- Cell Division
- Cells, Cultured
- DNA/analysis
- DNA/biosynthesis
- Diabetes Mellitus, Type 2/metabolism
- Disease Models, Animal
- Fibronectins/analysis
- Fibronectins/metabolism
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Platelet-Derived Growth Factor/analysis
- Platelet-Derived Growth Factor/pharmacology
- Proto-Oncogene Proteins c-sis
- Rats
- Rats, Inbred OLETF
- Receptor, Platelet-Derived Growth Factor beta/analysis
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Reference Values
- Sensitivity and Specificity
- Tunica Intima/injuries
- Tunica Intima/pathology
Collapse
Affiliation(s)
- K Tamura
- Second Department of Internal Medicine, School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, Japan
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Platelet-derived growth factor (PDGF) is a major mitogen for connective tissue cells and certain other cell types. It is a dimeric molecule consisting of disulfide-bonded, structurally similar A- and B-polypeptide chains, which combine to homo- and heterodimers. The PDGF isoforms exert their cellular effects by binding to and activating two structurally related protein tyrosine kinase receptors, denoted the alpha-receptor and the beta-receptor. Activation of PDGF receptors leads to stimulation of cell growth, but also to changes in cell shape and motility; PDGF induces reorganization of the actin filament system and stimulates chemotaxis, i.e., a directed cell movement toward a gradient of PDGF. In vivo, PDGF has important roles during the embryonic development as well as during wound healing. Moreover, overactivity of PDGF has been implicated in several pathological conditions. The sis oncogene of simian sarcoma virus (SSV) is related to the B-chain of PDGF, and SSV transformation involves autocrine stimulation by a PDGF-like molecule. Similarly, overproduction of PDGF may be involved in autocrine and paracrine growth stimulation of human tumors. Overactivity of PDGF has, in addition, been implicated in nonmalignant conditions characterized by an increased cell proliferation, such as atherosclerosis and fibrotic conditions. This review discusses structural and functional properties of PDGF and PDGF receptors, the mechanism whereby PDGF exerts its cellular effects, and the role of PDGF in normal and diseased tissues.
Collapse
Affiliation(s)
- C H Heldin
- Ludwig Institute for Cancer Research, Biomedical Center, and Department of Pathology, University Hospital, Uppsala, Sweden.
| | | |
Collapse
|
36
|
Masuda H, Goto M, Tamaoki S, Azuma H. Accelerated intimal hyperplasia and increased endogenous inhibitors for NO synthesis in rabbits with alloxan-induced hyperglycaemia. Br J Pharmacol 1999; 126:211-8. [PMID: 10051138 PMCID: PMC1565802 DOI: 10.1038/sj.bjp.0702298] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
1. We examined whether endogenous inhibitors of NO synthesis are involved in the augmentation of intimal hyperplasia in rabbits with hyperglycaemia induced by alloxan. 2. Four weeks after the endothelial denudation of carotid artery which had been performed 12 weeks after alloxan, the intimal hyperplasia was greatly augmented with hyperglycaemia. The degree of hyperplasia was assessed using three different parameters of histopathological findings as well as changes in luminal area and intima: media ratio. 3. There were positive and significant correlations between intima:media ratio, plasma glucose, and concentrations of N(G)-monomethyl-L-arginine (L-NMMA) and N(G), N(G)-dimethyl-L-arginine (ADMA) in endothelial cells, that is, the intima:media ratio became greater as plasma glucose and endothelial L-NMMA and ADMA were increased. Furthermore, endothelial L-NMMA and ADMA were increased in proportion to the increase in plasma glucose. 4. In contrast, there were inverse and significant correlations between cyclic GMP production by carotid artery strips with endothelium and plasma glucose, between cyclic GMP production and endothelial L-NMMA and ADMA, and between the intima:media ratio and cyclic GMP production. 5. Exogenously applied L-NMMA and ADMA inhibited cyclic GMP production in a concentration-dependent manner. IC50 values were determined to be 12.1 microM for the former and 26.2 microM for the latter. The cyclic GMP production was abolished after the deliberate removal of endothelium from the artery strips. 6. These results suggest that the augmentation of intimal hyperplasia with hyperglycaemia is closely related to increased accumulation of L-NMMA and ADMA with hyperglycaemia, which would result in an accelerated reduction in NO production/release by endothelial cells.
Collapse
Affiliation(s)
- Hiroshi Masuda
- Department of Medicinal Chemistry, Institute for Medical and Dental Engineering, Tokyo Medical and Dental University, 2-3-10 Surugadai, Kanda, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Moritaka Goto
- Department of Medicinal Chemistry, Institute for Medical and Dental Engineering, Tokyo Medical and Dental University, 2-3-10 Surugadai, Kanda, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Satoru Tamaoki
- Department of Medicinal Chemistry, Institute for Medical and Dental Engineering, Tokyo Medical and Dental University, 2-3-10 Surugadai, Kanda, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Hiroshi Azuma
- Department of Medicinal Chemistry, Institute for Medical and Dental Engineering, Tokyo Medical and Dental University, 2-3-10 Surugadai, Kanda, Chiyoda-ku, Tokyo 101-0062, Japan
- Author for correspondence:
| |
Collapse
|
37
|
Tamura K, Yokote K, Takemoto M, Matsumoto T, Ishisaki A, Funa K, Saito Y, Mori S. Fibronectin stimulates transcription of the platelet-derived growth factor beta-receptor in cultured rat aortic smooth muscle cells. Biochem Biophys Res Commun 1998; 251:677-80. [PMID: 9790968 DOI: 10.1006/bbrc.1998.9529] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fibronectin seems to play an important role in promoting the characteristic changes of vascular smooth muscle cells in diabetes mellitus including overexpression of the platelet-derived growth factor beta-receptor. To determine the regulatory mechanism of the beta-receptor by fibronectin, we have analyzed the effect of fibronectin on the expression of the beta-receptor in cultured rat aortic smooth muscle cells using the beta-receptor promoter/luciferase expression vector system. Fibronectin was found to stimulate the expression of the beta-receptor at the transcriptional level. Both a MEK1 inhibitor PD98059 and a tyrosine kinase inhibitor herbimycin A significantly inhibited the fibronectin-stimulated receptor transcription. Herbimycin A also completely inhibited the fibronectin-stimulated increase in tyrosine phosphorylation of focal adhesion kinase. These data suggest the involvement of the integrin-mediated mitogen-activated protein kinase pathway downstream of fibronectin stimulation in the activation process of the beta-receptor promoter.
Collapse
MESH Headings
- Animals
- Aorta/cytology
- Aorta/drug effects
- Benzoquinones
- Calcium-Calmodulin-Dependent Protein Kinases/metabolism
- Dose-Response Relationship, Drug
- Fibronectins/pharmacology
- Flavonoids/pharmacology
- Genes, Reporter
- Integrins/metabolism
- Lactams, Macrocyclic
- Luciferases/biosynthesis
- Luciferases/genetics
- Muscle, Smooth, Vascular/drug effects
- Promoter Regions, Genetic
- Protein Kinase Inhibitors
- Quinones/pharmacology
- Rats
- Receptor, Platelet-Derived Growth Factor beta
- Receptors, Platelet-Derived Growth Factor/biosynthesis
- Receptors, Platelet-Derived Growth Factor/genetics
- Recombinant Fusion Proteins/biosynthesis
- Rifabutin/analogs & derivatives
- Signal Transduction
- Transcription, Genetic
Collapse
Affiliation(s)
- K Tamura
- Second Department of Internal Medicine, School of Medicine, Chiba University, 1-8-1 Inohana, Chiba, Chuou-ku, 260-0856, Japan
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Tashiro J, Takahashi K, Yokote K, Takahashi M, Inadera H, Kobayashi J, Kanzaki T, Murano S, Shinomiya M, Morisaki N, Saito Y. Administration of a small amount of lard enhances intimal thickening in the balloon catheter injury model without affecting serum lipids. Scand J Clin Lab Invest 1998; 58:149-54. [PMID: 9587167 DOI: 10.1080/00365519850186733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Effects of fatty acids on intimal thickening induced by a balloon catheter injury model were investigated by feeding rabbits a small amount of either lard [L] or fish oil [F]. Serum lipids of these groups were not different from those of basal diet-fed rabbits [controls] after 4 weeks of feeding. Serum saturated fatty acids such as 14:0, 16:0, and 18.0 were significantly greater in the L-fed rabbits compared with controls, but those of the aorta were not significantly different. Fatty acid composition of the F-fed rabbits was only different from that of the controls in that n-3 fatty acids slightly increased. The mean and maximum intimal thickening 2 weeks after ballooning, carried out 2 weeks after feeding, were significantly higher in the carotid arteries of the L-fed rabbits than in the controls. The intimal thickening of the F-fed rabbits did not significantly differ from that of the controls. These results suggest that lard promotes the formation of the smooth muscle cell dominant type of arteriosclerosis without affecting serum lipid levels.
Collapse
Affiliation(s)
- J Tashiro
- Second Department of Internal Medicine, School of Medicine, Chiba University, Saiseikai-Funabashi Hospital, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Kornowski R, Mintz GS, Kent KM, Pichard AD, Satler LF, Bucher TA, Hong MK, Popma JJ, Leon MB. Increased restenosis in diabetes mellitus after coronary interventions is due to exaggerated intimal hyperplasia. A serial intravascular ultrasound study. Circulation 1997; 95:1366-9. [PMID: 9118501 DOI: 10.1161/01.cir.95.6.1366] [Citation(s) in RCA: 326] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND The increased risk of restenosis after catheter-based coronary interventions in diabetic patients has not been determined. Intravascular ultrasound (IVUS) has shown that the decrease in arterial area is responsible for most of the late lumen loss in nonstented lesions and that intimal hyperplasia is responsible for all of the late lumen loss in stented lesions. METHODS AND RESULTS Serial (postintervention and follow-up at 5.6 +/- 3.3 months) IVUS was used to study 251 native coronary lesions in 241 patients; 63 patients had treated diabetes mellitus (oral hypoglycemic drugs or insulin). Interventional procedures included percutaneous transluminal coronary angioplasty, directional or rotational atherectomy, excimer laser angioplasty, or Palmaz-Schatz stents. The external elastic membrane (EEM), stent, and lumen areas were measured. The plaque+media (P+M) area in nonstented lesions was calculated as EEM minus lumen area, and the intimal hyperplasia (IH) area in stented lesions was calculated as stent minus lumen area. The anatomic slice selected for serial analysis had an axial location within the target lesion at the smallest follow-up lumen area. Nonstented lesions in diabetics and nondiabetics had a similar decrease in EEM cross-sectional area (CSA; 1.9 +/- 2.8 versus 1.8 +/- 4.2 mm2; P = .6350). However, nonstented lesions in diabetics had a greater increase in P+M CSA (1.3 +/- 2.8 versus 0.6 +/- 2.5 mm2, P = .0720), and the increase in P+M CSA contributed a greater percentage to the decrease in lumen CSA. In stented lesions, the decrease in lumen CSA (5.2 +/- 2.5 versus 2.0 +/- 2.3 mm2) and the increase in IH CSA (5.0 +/- 2.8 versus 1.8 +/- 2.0 mm2) were greater in diabetics than nondiabetics (P = .0009 and P = .0007, respectively). These findings were even more striking in (nonstented and stented) restenotic lesions. CONCLUSIONS Serial IVUS analysis showed that the main reason for increased restenosis in diabetes mellitus was exaggerated intimal hyperplasia in both stented and nonstented lesions.
Collapse
Affiliation(s)
- R Kornowski
- Intravascular Imaging and Cardiac Catheterization Laboratories, Washington (DC) Hospital Center, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Diabetes is associated with greater restenosis rates after successful balloon angioplasty. The metabolic alterations that occur as a result of hyperglycemia or hyperinsulinemia can accelerate many of the pathophysiologic processes that lead to restenosis. Diabetes results in endothelial dysfunction and accelerated platelet deposition, which increase the propensity to thrombosis. Several growth factors known to promote the restenosis process are overexpressed in the presence of hyperglycemia. Advanced glycosylation promotes inflammatory cell recruitment and smooth muscle cell proliferation. Many of the potential mechanisms promoting restenosis in diabetic patients can be ameliorated by improved metabolic control.
Collapse
Affiliation(s)
- D Aronson
- Department of Medicine, Mount Sinai Medical Center, New York, NY 10029, USA
| | | | | |
Collapse
|
41
|
Kanzaki T, Tamura K, Takahashi K, Saito Y, Akikusa B, Oohashi H, Kasayuki N, Ueda M, Morisaki N. In vivo effect of TGF- beta 1. Enhanced intimal thickening by administration of TGF- beta 1 in rabbit arteries injured with a balloon catheter. Arterioscler Thromb Vasc Biol 1995; 15:1951-7. [PMID: 7583576 DOI: 10.1161/01.atv.15.11.1951] [Citation(s) in RCA: 85] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The in vivo effect of transforming growth factor-beta 1 (TGF-beta 1) was studied in a model system in which arterial intimal thickening was induced by injury of rabbit arteries with a balloon catheter (BCI). Intimal area and its ratio to medial area in carotid arteries after BCI were significantly higher in rabbits treated with 10 micrograms/kg TGF-beta 1 and 10 mg/kg aspirin i.v. QD (TGF-beta 1 group) than in those treated with 10 mg/kg aspirin i.v. QD only (control group). Intimal cell numbers in the TGF-beta 1 and control groups were not significantly different from each other, but matrix volume in the intimal layer was significantly higher in the TGF-beta 1 group. By immunohistochemical and Northern blot analyses, the fibronectin content in carotid intimal and medial layers was greater in the TGF-beta 1 group compared with that in the control group. Thus, in intimal thickenings induced by BCI. TGF-beta 1 mainly enhanced the formation of matrix containing fibronectin. Moreover, the mRNAs of TGF-beta 1 and type II receptors were detected in carotid arteries 7 and 14 days after, but not before, BCI. Thus, TGF-beta 1 influences the process of intimal thickening induced by BCI through a receptor-mediated mechanism in vivo. The significance of this fact is discussed in relation to the development of atherosclerosis.
Collapse
Affiliation(s)
- T Kanzaki
- Second Department of Internal Medicine, School of Medicine, Chiba University, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|