1
|
Dakroub A, Dbouk A, Asfour A, Nasser SA, El-Yazbi AF, Sahebkar A, Eid AA, Iratni R, Eid AH. C-peptide in diabetes: A player in a dual hormone disorder? J Cell Physiol 2024; 239:e31212. [PMID: 38308646 DOI: 10.1002/jcp.31212] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/05/2024]
Abstract
C-peptide, a byproduct of insulin synthesis believed to be biologically inert, is emerging as a multifunctional molecule. C-peptide serves an anti-inflammatory and anti-atherogenic role in type 1 diabetes mellitus (T1DM) and early T2DM. C-peptide protects endothelial cells by activating AMP-activated protein kinase α, thus suppressing the activity of NAD(P)H oxidase activity and reducing reactive oxygen species (ROS) generation. It also prevents apoptosis by regulating hyperglycemia-induced p53 upregulation and mitochondrial adaptor p66shc overactivation, as well as reducing caspase-3 activity and promoting expression of B-cell lymphoma-2. Additionally, C-peptide suppresses platelet-derived growth factor (PDGF)-beta receptor and p44/p42 mitogen-activated protein (MAP) kinase phosphorylation to inhibit vascular smooth muscle cells (VSMC) proliferation. It also diminishes leukocyte adhesion by virtue of its capacity to abolish nuclear factor kappa B (NF-kB) signaling, a major pro-inflammatory cascade. Consequently, it is envisaged that supplementation of C-peptide in T1DM might ameliorate or even prevent end-organ damage. In marked contrast, C-peptide increases monocyte recruitment and migration through phosphoinositide 3-kinase (PI-3 kinase)-mediated pathways, induces lipid accumulation via peroxisome proliferator-activated receptor γ upregulation, and stimulates VSMC proliferation and CD4+ lymphocyte migration through Src-kinase and PI-3K dependent pathways. Thus, it promotes atherosclerosis and microvascular damage in late T2DM. Indeed, C-peptide is now contemplated as a potential biomarker for insulin resistance in T2DM and linked to increased coronary artery disease risk. This shift in the understanding of the pathophysiology of diabetes from being a single hormone deficiency to a dual hormone disorder warrants a careful consideration of the role of C-peptide as a unique molecule with promising diagnostic, prognostic, and therapeutic applications.
Collapse
Affiliation(s)
- Ali Dakroub
- St. Francis Hospital and Heart Center, Roslyn, New York, USA
| | - Ali Dbouk
- Department of Medicine, Saint-Joseph University Medical School, Hotel-Dieu de France Hospital, Beirut, Lebanon
| | - Aref Asfour
- Leeds Teaching Hospitals NHS Trust, West Yorkshire, United Kingdom
| | | | - Ahmed F El-Yazbi
- Faculty of Pharmacy, Alamein International University (AIU), Alamein City, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, UAE
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
2
|
Yin J, Fu X, Luo Y, Leng Y, Ao L, Xie C. A Narrative Review of Diabetic Macroangiopathy: From Molecular Mechanism to Therapeutic Approaches. Diabetes Ther 2024; 15:585-609. [PMID: 38302838 PMCID: PMC10942953 DOI: 10.1007/s13300-024-01532-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/11/2024] [Indexed: 02/03/2024] Open
Abstract
Diabetic macroangiopathy, a prevalent and severe complication of diabetes mellitus, significantly contributes to the increased morbidity and mortality rates among affected individuals. This complex disorder involves multifaceted molecular mechanisms that lead to the dysfunction and damage of large blood vessels, including atherosclerosis (AS) and peripheral arterial disease. Understanding the intricate pathways underlying the development and progression of diabetic macroangiopathy is crucial for the development of effective therapeutic interventions. This review aims to shed light on the molecular mechanism implicated in the pathogenesis of diabetic macroangiopathy. We delve into the intricate interplay of chronic inflammation, oxidative stress, endothelial dysfunction, and dysregulated angiogenesis, all of which contribute to the vascular complications observed in this disorder. By exploring the molecular mechanism involved in the disease we provide insight into potential therapeutic targets and strategies. Moreover, we discuss the current therapeutic approaches used for treating diabetic macroangiopathy, including glycemic control, lipid-lowering agents, and vascular interventions.
Collapse
Affiliation(s)
- Jiacheng Yin
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Xiaoxu Fu
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, No. 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Yue Luo
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Yuling Leng
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Lianjun Ao
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, No. 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China.
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China.
| |
Collapse
|
3
|
Matwiejuk M, Myśliwiec H, Chabowski A, Flisiak I. An Overview of Growth Factors as the Potential Link between Psoriasis and Metabolic Syndrome. J Clin Med 2023; 13:109. [PMID: 38202116 PMCID: PMC10780265 DOI: 10.3390/jcm13010109] [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: 10/31/2023] [Revised: 11/26/2023] [Accepted: 12/05/2023] [Indexed: 01/12/2024] Open
Abstract
Psoriasis is a chronic, complex, and immunologically mediated systemic disease that not only affects the skin, but also the joints and nails. It may coexist with various other disorders, such as depression, psoriatic arthritis, cardiovascular diseases, diabetes mellitus, and metabolic syndrome. In particular, the potential link between psoriasis and metabolic syndrome is an issue worthy of attention. The dysregulation of growth factors could potentially contribute to the disturbances of keratinocyte proliferation, inflammation, and itch severity. However, the pathophysiology of psoriasis and its comorbidities, such as metabolic syndrome, remains incompletely elucidated. Growth factors and their abnormal metabolism may be a potential link connecting these conditions. Overall, the objective of this review is to analyze the role of growth factor disturbances in both psoriasis and metabolic syndrome.
Collapse
Affiliation(s)
- Mateusz Matwiejuk
- Department of Dermatology and Venereology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Hanna Myśliwiec
- Department of Dermatology and Venereology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Iwona Flisiak
- Department of Dermatology and Venereology, Medical University of Bialystok, 15-089 Bialystok, Poland
| |
Collapse
|
4
|
Sochett E, Noone D, Grattan M, Slorach C, Moineddin R, Elia Y, Mahmud FH, Dunger DB, Dalton N, Cherney D, Scholey J, Reich H, Deanfield J. Relationship between serum inflammatory markers and vascular function in a cohort of adolescents with type 1 diabetes. Cytokine 2017; 99:233-239. [PMID: 28760408 DOI: 10.1016/j.cyto.2017.07.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 06/27/2017] [Accepted: 07/17/2017] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The contribution of inflammation to endothelial/vascular dysfunction in early Type I Diabetes (T1D) is not well understood. The objective of this study was to examine the interaction between systemic inflammation and vascular function in adolescent's with and without-T1D. METHODS 51 subjects from our observational cohort of adolescents with T1D (JDRF-CCTN), and 59 healthy controls (HC) were studied. Serum cytokines-chemokines were quantified using Human 41-Plex Array, and vascular function was measured by Flow Mediated Dilatation (FMD), Pulse Wave Velocity (PWV) and Blood Pressure (BP). Factor Analysis was used to identify pro- and anti-inflammatory cytokine-chemokine factors, which were then correlated with vascular outcomes. RESULTS Three pro-inflammatory factors were identified in HC and three in TID, and a single anti-inflammatory factor in both groups. In HC there was a positive correlation (r=0.33; p=0.01) between control proinflammatory Factor 1 and systolic BP and a negative correlation between control proinflammatory Factor 3(r=-0.29; p=0.02) and diastolic BP. Control proinflammatory Factor 2 correlated positively with PWV. In TID subjects, no correlations were found between any of the pro-inflammatory factors and the vascular measurements. No correlations were found between the anti-inflammatory factors and BP, FMD and PWV in either HC or T1D. Levels of pro-inflammatory analytes, EGF, GRO, PDGF-BB, PDGF-AA and sCD40L were significantly higher in T1D. CONCLUSIONS The cytokine-chemokine signature in early T1D, prior to the development of arterial disease, is significantly different from that seen in healthy controls. This may be relevant to pathophysiology, determining risk and identifying target cytokines-chemokines for intervention in T1D.
Collapse
Affiliation(s)
- Etienne Sochett
- Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, Canada; Division of Endocrinology, Hospital for Sick Children, Toronto, Canada.
| | - Damien Noone
- Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, Canada; Division of Nephrology, Hospital for Sick Children, Toronto, Canada
| | - Michael Grattan
- Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, Canada; London Health Sciences Centre, Children's Hospital, University of Western Ontario, London, Ontario, Canada
| | - Cameron Slorach
- Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Rahim Moineddin
- Department of Family and Community Medicine, University of Toronto, Toronto, Canada
| | - Yesmino Elia
- Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Farid H Mahmud
- Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - David B Dunger
- Department of Paediatrics, University of Cambridge, Cambridge, UK Department of Medicine, UK
| | - Neil Dalton
- WellChild Laboratory, Evelina Children's Hospital, St Thomas' Hospital, London, UK
| | - David Cherney
- Division of Nephrology, University Health Network, University of Toronto, Toronto, Canada
| | - James Scholey
- Division of Nephrology, University Health Network, University of Toronto, Toronto, Canada
| | - Heather Reich
- Division of Nephrology, University Health Network, University of Toronto, Toronto, Canada
| | - John Deanfield
- Institute of Child Health, University College London, London, UK
| |
Collapse
|
5
|
Ouyang L, Zhang K, Chen J, Wang J, Huang H. Roles of platelet-derived growth factor in vascular calcification. J Cell Physiol 2017; 233:2804-2814. [PMID: 28467642 DOI: 10.1002/jcp.25985] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/02/2017] [Indexed: 02/06/2023]
Abstract
Vascular calcification (VC) is prevalent in aging, and patients with hypertension, chronic kidney disease (CKD), or diabetes. VC is regarded as an active and complex process that involves multiple mechanisms responsible for calcium deposition in vessel wall. In light of the complicated pathogenesis of VC, effective therapy for ameliorating VC is limited. Thus, it is urgent to explore the potential mechanisms and find new targets for the therapy of VC. Platelet-derived growth factor (PDGF), a potent mitogen, and chemoattractant have been found to disturb the vascular homeostasis by inducing inflammation, oxidative stress, and phenotype transition, all of which accelerate the process of VC. The aim of current review is to present a review about the roles of PDGF in affecting VC and to establish a potential target for treating VC.
Collapse
Affiliation(s)
- Liu Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China
| | - Kun Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China
| | - Jie Chen
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China.,Department of Radiation Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jingfeng Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China
| | - Hui Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China
| |
Collapse
|
6
|
Hu W, Huang Y. Targeting the platelet-derived growth factor signalling in cardiovascular disease. Clin Exp Pharmacol Physiol 2015; 42:1221-4. [DOI: 10.1111/1440-1681.12478] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 08/03/2015] [Accepted: 08/08/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Weining Hu
- Shenzhen Research Institute; Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences; Chinese University of Hong Kong; Hong Kong China
| | - Yu Huang
- Shenzhen Research Institute; Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences; Chinese University of Hong Kong; Hong Kong China
| |
Collapse
|
7
|
Prasad K. Pathophysiology and Medical Treatment of Carotid Artery Stenosis. Int J Angiol 2015; 24:158-72. [PMID: 26417183 DOI: 10.1055/s-0035-1554911] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Stroke is the third leading cause of mortality. Approximately 80 to 85% strokes are ischemic due to carotid artery stenosis (CAS). The prevalence of significant CAS is 7% in women and 9% in men. Severe asymptomatic CAS varies from 0 to 3.1%. Prevalence of symptomatic CAS is high in patients with peripheral arterial disease. CAS is due to atherosclerosis, the major risk factors for which include dyslipidemia, hypertension, diabetes, obesity, cigarette smoking, advanced glycation end products (AGEs) and its receptors (RAGE, soluble RAGE [sRAGE]), lack of exercise and C-reactive protein (CRP). This article discusses the basic mechanism of atherosclerosis and the mechanisms by which these risk factors induce atherosclerosis. The role of AGEs and its receptors in the development and progression of CAS has been discussed in detail. Lifestyle changes and medical treatment of CAS such as lifestyle changes, lipid-lowering agents, antihypertensive agents, antidiabetic drugs, anti-AGE therapy, measures to elevate soluble receptors of AGE (sRAGE, esRAGE). CRP-lowering agents have been discussed in detail. The drugs especially lipid-lowering agents, and antihypertensive and antidiabetic drugs suppress, regress, and slow the progression of CAS. The possible role of lowering the levels of AGEs and raising the levels of sRAGE in the treatment of CAS has been proposed. Lifestyle changes besides medical treatment have been stressed. Lifestyle changes and medical treatment not only would slow the progression of CAS but would also regress the CAS.
Collapse
Affiliation(s)
- Kailash Prasad
- Department of Physiology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
8
|
Abstract
The rising epidemic of T2DM (Type 2 diabetes mellitus) worldwide is of significant concern. The inherently silent nature of the disease in its early stages precludes early detection; hence cardiovascular disease is often established by the time diabetes is diagnosed. This increased cardiovascular risk leads to significant morbidity and mortality in these individuals. Progressive development of complications as a result of previous exposure to metabolic disturbances appears to leave a long-lasting impression on cells of the vasculature that is not easily reversed and is termed 'metabolic memory'. SMCs (smooth muscle cells) of blood vessel walls, through their inherent ability to switch between a contractile quiescent phenotype and an active secretory state, maintain vascular homoeostasis in health and development. This plasticity also confers SMCs with the essential capacity to adapt and remodel in pathological states. Emerging clinical and experimental studies propose that SMCs in diabetes may be functionally impaired and thus contribute to the increased incidence of macrovascular complications. Although this idea has general support, the underlying molecular mechanisms are currently unknown and hence are the subject of intense research. The aim of the present review is to explore and evaluate the current literature relating to the problem of vascular disease in T2DM and to discuss the critical role of SMCs in vascular remodelling. Possibilities for therapeutic strategies specifically at the level of T2DM SMCs, including recent novel advances in the areas of microRNAs and epigenetics, will be evaluated. Since restoring glucose control in diabetic patients has limited effect in ameliorating their cardiovascular risk, discovering alternative strategies that restrict or reverse disease progression is vital. Current research in this area will be discussed.
Collapse
|
9
|
Hu P, Lai D, Lu P, Gao J, He H. ERK and Akt signaling pathways are involved in advanced glycation end product-induced autophagy in rat vascular smooth muscle cells. Int J Mol Med 2012; 29:613-8. [PMID: 22293957 PMCID: PMC3573741 DOI: 10.3892/ijmm.2012.891] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 12/29/2011] [Indexed: 12/14/2022] Open
Abstract
Advanced glycation end products (AGEs) play an important role in the proliferation of vascular smooth muscle cells (VSMCs) and accelerate atherosclerosis in diabetic patients. Autophagy, a life-sustaining process, is stimulated in atherosclerotic plaques by oxidized lipids, inflammation and metabolic stress conditions. In our studies, we utilized MTT assays to show that autophagy is involved in AGE-induced proliferation of VSMCs. Furthermore, treatment with AGEs (100 μg/ml) could induce autophagy in a time- and dose-dependent manner in rat aortic VSMCs. These results were further substantiated by electron microscopy and immunofluorescence imaging. Treatment with AGEs activated ERK, JNK and p38/MAPK, but inhibited Akt. Pretreatment with an ERK inhibitor and an Akt activator inhibited AGE-induced autophagy, demonstrating that AGEs induce autophagy in VSMCs through the ERK and Akt signaling pathways. In addition, RNA interference of RAGE decreased autophagy, indicating that RAGE is pivotal in the process of AGE-induced autophagy. Therefore, AGE-induced autophagy contributes to the process of AGE-induced proliferation of VSMCs, which is related to atherosclerosis in diabetes.
Collapse
Affiliation(s)
- Pengfei Hu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang Province, PR China
| | | | | | | | | |
Collapse
|
10
|
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: 2.2] [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]
|
11
|
Swiderek M, Kozubski W, Watala C. Abnormalities in platelet membrane structure and function in Alzheimer's disease and ischaemic stroke. Platelets 2010; 8:125-33. [DOI: 10.1080/09537109709169327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
12
|
Descorbeth M, Anand-Srivastava MB. Role of growth factor receptor transactivation in high glucose-induced increased levels of Gq/11alpha and signaling in vascular smooth muscle cells. J Mol Cell Cardiol 2009; 49:221-33. [PMID: 20036247 DOI: 10.1016/j.yjmcc.2009.12.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Accepted: 12/14/2009] [Indexed: 11/29/2022]
Abstract
We have recently shown that high glucose increased the expression of Gq/11alpha, PLCbeta and mediated signaling in A10 vascular smooth muscle cells (VSMC). Since high glucose has been shown to increase growth factor receptor activation, we investigated the role of epidermal growth factor receptor (EGF-R) and platelet-derived growth factor receptor (PDGF-R) transactivation in high glucose-induced enhanced expression of Gq/11alpha and PLCbeta. Pre-treatment of A10 VSMC with high glucose (26 mM) for 3 days, increased the levels of Gqalpha, G11alpha, PLCbeta-1 and PLCbeta-2 proteins which were restored to control levels by AG1478, an inhibitor of EGF-R, AG1295, an inhibitor of PDGF-R and PP2, an inhibitor of c-Src but not by PP3. In addition, endothelin-1 (ET-1)-stimulated production of IP(3) that was enhanced by high glucose was also restored towards control levels by AG1478, AG1295 and PP2. High glucose also increased the phosphorylation of EGF-R and PDGF-R which was abolished by AG1478, AG1295 and PP2. Furthermore, high glucose-induced enhanced levels of Gqalpha, G11alpha and PLCbeta were also attenuated by PD98059, an inhibitor of mitogen-activated protein kinase (MAPK) and wortmannin, an inhibitor of phosphatidylinositol 3-kinase (PI3-K). In addition, AG1478 and AG1295, also attenuated high glucose-induced enhanced phosphorylation of ERK1/2 and AKT. Furthermore, high glucose augmented the phosphorylation of c-Src which was attenuated by antioxidant, DPI. These results suggest that oxidative stress through the activation of c-Src and resultant transactivation of growth factor receptor contributes to the high glucose-induced enhanced expression of Gq/11alpha/PLC and -mediated cell signaling through MAPK/PI3K pathway.
Collapse
Affiliation(s)
- Magda Descorbeth
- Department of Physiology, University of Montreal, Montreal, Quebec, Canada
| | | |
Collapse
|
13
|
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]
|
14
|
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]
|
15
|
Campbell M, Anderson P, Trimble ER. Glucose lowers the threshold for human aortic vascular smooth muscle cell migration: inhibition by protein phosphatase-2A. Diabetologia 2008; 51:1068-80. [PMID: 18340431 DOI: 10.1007/s00125-008-0962-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2007] [Accepted: 01/11/2008] [Indexed: 10/22/2022]
Abstract
AIMS/HYPOTHESIS Atherosclerosis, which occurs prematurely in individuals with diabetes, incorporates vascular smooth muscle cell (VSMC) chemotaxis. Glucose, through protein kinase C-beta(II) signalling, increases chemotaxis to low concentrations of platelet-derived growth factor (PDGF)-BB. In VSMC, a biphasic response in PDGF-beta receptor (PDGF-betaR) level occurs as PDGF-BB concentrations increase. The purpose of this study was to determine whether increased concentrations of PDGF-BB and raised glucose level had a modulatory effect on the mitogen-activated protein kinase/extracellular-regulated protein kinase pathway, control of PDGF-betaR level and chemotaxis. METHODS Cultured aortic VSMC, exposed to normal glucose (NG) (5 mmol/l) or high glucose (HG) (25 mmol/l) in the presence of PDGF-BB, were assessed for migration (chemotaxis chamber) or else extracted and immunoblotted. RESULTS At concentrations of PDGF-BB <540 pmol/l, HG caused an increase in the level of PDGF-betaR in VSMC (immunoblotting) versus NG, an effect that was abrogated by inhibition of aldose reductase or protein kinase C-beta(II). At higher concentrations of PDGF-BB (>540 pmol/l) in HG, receptor level was reduced but in the presence of aldose reductase or protein kinase C-beta(II) inhibitors the receptor levels increased. It is known that phosphatases may be activated at high concentrations of growth factors. At high concentrations of PDGF-BB, the protein phosphatase (PP)2A inhibitor, endothall, caused an increase in PDGF-betaR levels and a loss of biphasicity in receptor levels in HG. At higher concentrations of PDGF-BB in HG, the chemoattractant effect of PDGF-BB was lost (chemotaxis chamber). Under these conditions inhibition of PP2A was associated with a restoration of chemotaxis to high concentrations of PDGF-BB. CONCLUSION/INTERPRETATION The biphasic response in PDGF-betaR level and in chemotaxis to PDGF-BB in HG is due to PP2A activation.
Collapse
Affiliation(s)
- M Campbell
- Diabetes Research Group, Queen's University Belfast, Grosvenor Road, Belfast BT12 6BJ, UK.
| | | | | |
Collapse
|
16
|
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: 1.6] [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
|
17
|
Rahman S, Rahman T, Ismail AAS, Rashid ARA. Diabetes-associated macrovasculopathy: pathophysiology and pathogenesis. Diabetes Obes Metab 2007; 9:767-80. [PMID: 17924861 DOI: 10.1111/j.1463-1326.2006.00655.x] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The complications associated with diabetic vasculopathy are commonly grouped into two categories: microvascular and macrovascular complications. In diabetes, macrovascular disease is the commonest cause of mortality and morbidity and is responsible for high incidence of vascular diseases such as stroke, myocardial infarction and peripheral vascular diseases. Macrovascular diseases are traditionally thought of as due to underlying obstructive atherosclerotic diseases affecting major arteries. Pathological changes of major blood vessels leading to functional and structural abnormalities in diabetic vessels include endothelial dysfunction, reduced vascular compliance and atherosclerosis. Besides, advanced glycation end product formation interacts with specific receptors that lead to overexpression of a range of cytokines. Haemodynamic pathways are activated in diabetes and are possibly amplified by concomitant systemic hypertension. Apart from these, hyperglycaemia, non-enzymatic glycosylation, lipid modulation, alteration of vasculature and growth factors activation contribute to development of diabetic vasculopathy. This review focuses on pathophysiology and pathogenesis of diabetes-associated macrovasculopathy.
Collapse
Affiliation(s)
- Sayeeda Rahman
- Department of Pharmacology, School of Medical Sciences, Kubang Kerian, University Sains Malaysia, Kelantan, Malaysia.
| | | | | | | |
Collapse
|
18
|
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: 80] [Impact Index Per Article: 4.4] [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
|
19
|
Kobayashi Y, Naruse K, Hamada Y, Nakashima E, Kato K, Akiyama N, Kamiya H, Watarai A, Nakae M, Oiso Y, Nakamura J. Human proinsulin C-peptide prevents proliferation of rat aortic smooth muscle cells cultured in high-glucose conditions. Diabetologia 2005; 48:2396-401. [PMID: 16195866 DOI: 10.1007/s00125-005-1942-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2004] [Accepted: 06/29/2005] [Indexed: 12/01/2022]
Abstract
AIMS/HYPOTHESIS Proinsulin C-peptide is involved in several biological activities. However, the role of C-peptide in vascular smooth muscle cells is unclear. We therefore investigated its effects, in vascular smooth muscle cells in high-glucose conditions. METHODS Rat aortic smooth muscle cells were cultured with 5.5 or 20 mmol/l glucose with or without C-peptide (1 to 100 nmol/l) for 3 weeks. Proliferation activities, the protein expression of platelet-derived growth factor (PDGF)-beta receptor, the phosphorylation of p42/p44 mitogen-activated protein (MAP) kinases, and glucose uptake were measured. RESULTS The proliferation activities increased approximately three-fold under high-glucose conditions (p<0.05). C-peptide suppressed hyperproliferation activities that were induced by high glucose. This happened in a dose-dependent manner from 1 to 100 nmol/l of C-peptide. C-peptide (10 and 100 nmol/l) inhibited the increased protein expression of PDGF-beta receptor and the phosphorylation of p42/p44 MAP kinases that had been induced by high glucose (p<0.05). Furthermore, 100 nmol/l of C-peptide augmented the impaired glucose uptake in the high-glucose conditions. CONCLUSIONS/INTERPRETATION These observations suggest that C-peptide could prevent diabetic macroangiopathy by inhibiting smooth muscle cell growth and ameliorating glucose utilisation in smooth muscle cells. C-peptide may thus be a novel agent for treating diabetic macroangiopathy in patients with type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Y Kobayashi
- Division of Metabolic Diseases, Department of Internal Medicine, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
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
|
21
|
Campbell M, Trimble ER. Modification of PI3K- and MAPK-Dependent Chemotaxis in Aortic Vascular Smooth Muscle Cells by Protein Kinase C
βII. Circ Res 2005; 96:197-206. [PMID: 15591231 DOI: 10.1161/01.res.0000152966.88353.9d] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Hyperglycemia increases expression of platelet-derived growth factor (PDGF)-β receptor and potentiates chemotaxis to PDGF-BB in human aortic vascular smooth muscle cells (VSMCs) via PI3K and ERK/MAPK signaling pathways. The purpose of this study was to determine whether increased activation of protein kinase C (PKC) isoforms had a modulatory effect on the PI3K and ERK/MAPK pathways, control of cell adhesiveness, and movement. All known PKC isoforms were assessed but only PKC
α
and PKC
βII
levels were increased in 25 mmol/L glucose. However, only PKC
βII
inhibition affected (decreased) PI3K pathway and MAPK pathway activities and inhibited PDGF-β receptor upregulation in raised glucose, and specific MAPK inhibition was required to completely block the effect of glucose. In raised glucose conditions, activity of the ERK/MAPK pathway, PI3K pathway, and PKC
βII
were all sensitive to aldose reductase inhibition. Chemotaxis to PDGF-BB (360 pmol/L), absent in 5 mmol/L glucose, was present in raised glucose and could be blocked by PKC
βII
inhibition. Formation of lamellipodia was dependent on PI3K activation and filopodia on MAPK activation; both lamellipodia and filopodia were eliminated when PKC
βII
was inhibited. FAK phosphorylation and cell adhesion were reduced by PI3K inhibition, and although MAPK inhibition prevented chemotaxis, it did not affect FAK phosphorylation or cell adhesiveness. In conclusion, chemotaxis to PDGF-BB in 25 mmol/L glucose is PKC
βII
-dependent and requires activation of both the PI3K and MAPK pathways. Changes in cell adhesion and migration speed are mediated mainly through the PI3K pathway.
Collapse
Affiliation(s)
- Malcolm Campbell
- Department of Clinical Biochemistry and Metabolic Medicine, Queen's University Belfast, Institute of Clinical Science, Royal Victoria Hospital, Grosvenor Rd, Belfast BT12 6BJ, UK.
| | | |
Collapse
|
22
|
Jeong HY, Son SM, Kim YK, Yun MR, Lee SM, Kim CD. Tyrosine kinase-mediated activation of NADPH oxidase enhances proliferative capacity of diabetic vascular smooth muscle cells. Life Sci 2005; 76:1747-57. [PMID: 15698853 DOI: 10.1016/j.lfs.2004.09.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2004] [Accepted: 09/28/2004] [Indexed: 11/21/2022]
Abstract
To investigate a potential molecular basis for a link between diabetes and atherosclerosis, experiments were performed to determine the role of NADPH oxidase in the enhanced proliferative capacity of vascular smooth muscle cells (VSMC) from OLETF rat, an animal model of type 2 diabetes. An enhanced proliferative response to 10% fetal bovine serum with an increased cell cycle progression from G1 to S phase as well as an augmented superoxide generation with an increased NADPH oxidase activity were observed in diabetic versus control VSMC. Both the enhanced proliferation and superoxide generation in diabetic VSMC were significantly attenuated not only by diphenyleneiodonium (10 microM) and apocynin (100 microM), NADPH oxidase inhibitors but also by protein tyrosine kinase inhibitors such as genistein (100 microM) and AG 112 (100 microM). Furthermore, the enhanced NADPH oxidase activity in diabetic VSMC was significantly attenuated by genistein and AG112, but not by daidzein (100 microM), a genistein analogue devoid of protein tyrosine kinase inhibitory properties. Based on these results, it is suggested that the enhanced proliferative capacity of diabetic VSMC is closely related to the activation of NADPH oxidase that is induced through activation of protein tyrosine kinase.
Collapse
Affiliation(s)
- Hye Young Jeong
- Department of Pharmacology and Internal Medicine, College of Medicine, Research Institute of Genetic Engineering and Medical Research Institute, Pusan National University, 10 Ami-Dong 1-Ga, Seo-Gu, Busan 602-739, South Korea
| | | | | | | | | | | |
Collapse
|
23
|
Ikeda Y, Suehiro T, Osaki F, Tsuzura S, Kumon Y, Hashimoto K. Polymorphisms in the 5'-upstream region of the PKCbeta gene in Japanese patients with Type 2 diabetes. Diabet Med 2004; 21:1113-20. [PMID: 15384959 DOI: 10.1111/j.1464-5491.2004.01304.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIMS Protein kinase C (PKC), a serine/threonine kinase, is known to be activated in various tissues under hyperglycaemic conditions. Notably, PKCbeta, a member of the conventional PKC group, is the predominant isoform detected in vascular tissues and could be involved in the development of diabetic vascular complications. In the present study, we investigated genetic variations in the 5'-upstream region of the PKCbeta gene to assess their possible relation to vascular complications in diabetic patients. METHODS Variations upstream from the PKCbeta gene (-1066/+256) were examined in 60 Type 2 diabetic patients using a cycle sequencing method. Screening of detected variations was performed in 204 Type 2 diabetic patients and 160 healthy controls. RESULTS Five single nucleotide polymorphisms; C(-238)G, C(-287)T, A(-348)G, C(-546)G, and C(-853)T, were identified in the upstream region. The C(-287)T and A(-348)G polymorphisms were in perfect linkage disequilibrium. There were no significant differences in genotype or allele frequencies of the five polymorphisms among the diabetic patients and healthy subjects. However, both -238GG and -287CC (-348GG) homozygotes showed significantly higher frequencies of macrovascular disease compared with patients with other genotypes. Further, an electrophoretic mobility shift assay revealed that the -238G fragment had a five-fold higher affinity for transcription factor Sp1 when compared with -238C. CONCLUSIONS The C(-238)G and C(-287)T-A(-348)G polymorphisms in the 5'-upstream region of the PKCbeta gene may have an effect on the susceptibility of diabetic vascular complications through an alteration of tissue PKCbeta density or function.
Collapse
Affiliation(s)
- Y Ikeda
- Department of Endocrinology, Metabolism and Nephrology, Kochi Medical School, Japan.
| | | | | | | | | | | |
Collapse
|
24
|
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: 2.1] [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
|
25
|
Jeong HY, Jeong HY, Kim CD. p22phox-derived superoxide mediates enhanced proliferative capacity of diabetic vascular smooth muscle cells. Diabetes Res Clin Pract 2004; 64:1-10. [PMID: 15036821 DOI: 10.1016/j.diabres.2003.10.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2003] [Revised: 07/14/2003] [Accepted: 10/08/2003] [Indexed: 11/23/2022]
Abstract
To investigate the mechanisms that contribute to the acceleration of atherosclerosis in diabetes, the role of NAD(P)H oxidase in the enhanced proliferative capacity of diabetic vascular smooth muscle cells (VSMC) was studied. VSMC from streptozotocin (STZ)-induced diabetic rat aorta had increased proliferative capacity and generated higher levels of superoxide in comparison with cells from control rats. Both the enhanced proliferation and superoxide generation in diabetic VSMC were significantly attenuated not only by tiron (1mM), a superoxide scavenger but also by diphenyleneiodonium (DPI; 10microM), an NAD(P)H oxidase inhibitor. Both the activity of NAD(P)H oxidase and p22phox expression were significantly increased in diabetic VSMC. Furthermore, inhibition of p22phox expression by transfection of antisense p22phox oligonucleotides into diabetic VSMC resulted in a decrease in superoxide generation, which was accompanied by a significant attenuation of cell proliferation. Based on these results, it is suggested that diabetes-associated increase in NAD(P)H oxidase activity via enhanced expression of p22phox contributes to augmented VSMC proliferation in diabetic rats.
Collapse
MESH Headings
- Animals
- Aorta/drug effects
- Aorta/pathology
- Base Sequence
- Cell Division/drug effects
- Cells, Cultured
- DNA Primers
- Diabetes Mellitus, Experimental/pathology
- Disease Models, Animal
- Gene Expression Regulation, Enzymologic/genetics
- Male
- Membrane Transport Proteins/genetics
- Membrane Transport Proteins/pharmacology
- Membrane Transport Proteins/physiology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- NAD/metabolism
- NADH, NADPH Oxidoreductases/genetics
- NADPH Dehydrogenase/genetics
- NADPH Dehydrogenase/pharmacology
- NADPH Dehydrogenase/physiology
- NADPH Oxidases
- Oligonucleotides, Antisense/pharmacology
- Phosphoproteins/genetics
- Phosphoproteins/pharmacology
- Phosphoproteins/physiology
- Rats
- Rats, Sprague-Dawley
- Reverse Transcriptase Polymerase Chain Reaction
- Superoxides/pharmacology
- Transfection
- Xanthine/metabolism
Collapse
Affiliation(s)
- Hye Y Jeong
- Department of Pharmacology, College of Medicine, Research Institute of Genetic Engineering, Pusan National University, 10 Ami-Dong 1-Ga, Seo-Gu, Busan 602-739, South Korea
| | | | | |
Collapse
|
26
|
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
|
27
|
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
|
28
|
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
|
29
|
Little PJ, Allen TJ, Hashimura K, Nigro J, Farrelly CA, Dilley RJ. High glucose potentiates mitogenic responses of cultured ovine coronary smooth muscle cells to platelet derived growth factor and transforming growth factor-beta1. Diabetes Res Clin Pract 2003; 59:93-101. [PMID: 12560158 DOI: 10.1016/s0168-8227(02)00201-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Macrovascular complications in diabetes are associated with exaggerated growth responses of vascular smooth muscle cells. We studied the effect of high glucose media on the growth responses of vascular smooth muscle cells from the left anterior descending (LAD) coronary artery of young sheep. Experiments were conducted in DMEM containing 5.5 or 25 mmol/l glucose and mitogenic responses assessed by 3H-thymidine incorporation. In the absence of growth factors there was a slight and variable response to high glucose but the maximum response to platelet derived growth factor-bb (PDGF-bb) (100 ng/ml) was increased more than 2-fold. Transforming growth factor-beta1 (1 ng/ml) caused a 100% increase of the PDGF-bb response in both normal and high glucose media. The acute stimulatory effect of high glucose was not affected by pre-incubation of the cells for 24 h in the high glucose medium. The mitogenic response occurring in the presence of PDGF-bb and high glucose was totally inhibited by the tyrosine kinase inhibitors (imatinib and genistein) and could not be mimicked by increasing diacylglycerol in low glucose media with the diacylglycerol kinase inhibitor, R59949. In conclusion, high glucose, per se, only very weakly stimulates smooth muscle cell growth but it interacts positively to potentiate the responses to the vascular derived growth factors PDGF and TGF-beta1. The effect of high glucose is transduced via receptor tyrosine kinases and may not involve diacylglycerol that is subject to diacylglycerol kinase catabolism. The data provide explanations for the accelerated vascular smooth muscle cell proliferation in diabetes.
Collapse
Affiliation(s)
- Peter J Little
- Cell Biology of Diabetes Laboratory, Baker Heart Research Institute and The Alfred Hospital, St. Kilda Road Central, PO Box 6492, Melbourne, Vic. 8008, Australia.
| | | | | | | | | | | |
Collapse
|
30
|
Igarashi M, Yamaguchi H, Hirata A, Tsuchiya H, Ohnuma H, Tominaga M, Daimon M, Kato T. Mechanisms of inhibitory effects of cerivastatin on rat vascular smooth muscle cell growth. J Cardiovasc Pharmacol 2002; 40:277-87. [PMID: 12131557 DOI: 10.1097/00005344-200208000-00013] [Citation(s) in RCA: 7] [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
The aim of this study was to clarify the mechanism(s) of an inhibitory effect of cerivastatin on cultured rat vascular smooth muscle cell (VSMC) growth. After being starved, cultured VSMCs were stimulated by 5% fetal bovine serum with either various concentrations of cerivastatin or 10-4 M of mevalonate. Cerivastatin dose-dependently decreased the values of [3H]-thymidine incorporation and cell numbers and the level of phosphorylated extracellular signal-regulated protein kinase 1/2. It also suppressed the level of proliferative cell nuclear antigen in a dose-dependent manner. These reductions were abolished by the addition of mevalonate. Similarly, the level of phosphorylated p38 was also decreased by cerivastatin. In contrast, cerivastatin dose-dependently activated the phosphorylation of both c-jun NH2-terminal protein kinase and activating transcription factor-2, and these activations were abolished by the addition of mevalonate. The levels of phosphorylated Akt and p70 S6 kinase as well as those of Bcl-2 were dose-dependently reduced by cerivastatin, and these reductions were abolished by the addition of mevalonate. Cerivastatin could dose-dependently elevate the levels of CPP32/caspase-3 activity and cytoplasmic histone-associated DNA fragments in VSMCs without causing cytotoxicity. These results indicate that cerivastatin suppresses cell survival and activates the apoptotic cellular signaling in VSMCs, suggesting that it could be effective for preventing the progression of restenosis after angioplasty.
Collapse
Affiliation(s)
- Masahiko Igarashi
- Third Department of Internal Medicine and Department of Laboratory Medicine, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Aronson D, Rayfield EJ. How hyperglycemia promotes atherosclerosis: molecular mechanisms. Cardiovasc Diabetol 2002; 1:1. [PMID: 12119059 PMCID: PMC116615 DOI: 10.1186/1475-2840-1-1] [Citation(s) in RCA: 314] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2002] [Accepted: 04/08/2002] [Indexed: 02/02/2023] Open
Abstract
Both type I and type II diabetes are powerful and independent risk factors for coronary artery disease (CAD), stroke, and peripheral arterial disease. Atherosclerosis accounts for virtually 80% of all deaths among diabetic patients. Prolonged exposure to hyperglycemia is now recognized a major factor in the pathogenesis of atherosclerosis in diabetes. Hyperglycemia induces a large number of alterations at the cellular level of vascular tissue that potentially accelerate the atherosclerotic process. Animal and human studies have elucidated three major mechanisms that encompass most of the pathological alterations observed in the diabetic vasculature: 1) Nonenzymatic glycosylation of proteins and lipids which can interfere with their normal function by disrupting molecular conformation, alter enzymatic activity, reduce degradative capacity, and interfere with receptor recognition. In addition, glycosylated proteins interact with a specific receptor present on all cells relevant to the atherosclerotic process, including monocyte-derived macrophages, endothelial cells, and smooth muscle cells. The interaction of glycosylated proteins with their receptor results in the induction of oxidative stress and proinflammatory responses 2) oxidative stress 3) protein kinase C (PKC) activation with subsequent alteration in growth factor expression. Importantly, these mechanisms may be interrelated. For example, hyperglycemia-induced oxidative stress promotes both the formation of advanced glycosylation end products and PKC activation.
Collapse
Affiliation(s)
- Doron Aronson
- Cardiology Division, Rambam Medical Center, 31096 Haifa, Israel
| | - Elliot J Rayfield
- Clinical Professor of Medicine Mount Sinai School of Medicine, New York, NY 10029, USA
- USA Visiting Physician, Rockefeller University Hospital, New York, NY 10021, USA
| |
Collapse
|
32
|
Wang R, Kudo M, Yokoyama M, Asano G. Roles of advanced glycation endproducts (AGE) and receptor for AGE on vascular smooth muscle cell growth. J NIPPON MED SCH 2001; 68:472-81. [PMID: 11744927 DOI: 10.1272/jnms.68.472] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Proliferation of vascular smooth muscle cells (VSMC) represents an essential event in the developement of diabetic atherosclerosis. Previous studies suggest that several cytokines and growth factors mediate the proliferation capability in VSMC from diabetic animals. In addition, advanced glycation end products (AGE) and receptor for AGE (RAGE) are important for pathologic features of diabetic complications. In the present study, we attempted to clarify the roles of AGE and RAGE in the proliferation of VSMC using streptozotocin (STZ)-treated rat sera and aortic SMC prepared from non-diabetic rats. AGE levels increased in the diabetic sera, which enhanced the growth of VSMC in proportion to their diabetic periods. AGE-bovine serum albumin (BSA) prepared in vitro also exhibited a stimulatory effect on VSMC growth. The endocytic uptake of AGE and enhanced RAGE expression in VSMC after culture with diabetic sera were observed. In addition, anti-AGE and anti-RAGE antibodies inhibited these stimulatory effects on VSMC growth. These findings suggest that AGE in diabetic rat sera may cause an enhanced effect on VSMC proliferation. However, the concentrations of AGE in diabetic sera were much lower than that of AGE-BSA which demonstrated a significant stimulatory effect on VSMC growth. The magnitude of the VSMC growth-enhancement by the diabetic sera was markedly greater than that by the AGE-BSA solution. In conclusion, the AGE-RAGE interaction in VSMC, in addition to growth factors induced by AGE, contributes to the stimulatory effect of diabetic sera on VSMC proliferation which can accelerate atherosclerosis.
Collapse
MESH Headings
- Animals
- Arteriosclerosis/etiology
- Cell Division
- Cells, Cultured
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/physiopathology
- Diabetic Angiopathies/etiology
- Glycation End Products, Advanced/metabolism
- Glycation End Products, Advanced/physiology
- Male
- Muscle, Smooth, Vascular/cytology
- Rats
- Rats, Wistar
- Receptor for Advanced Glycation End Products
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/physiology
- Streptozocin
Collapse
Affiliation(s)
- R Wang
- Department of Pathology, Nippon Medical School, Tokyo, Japan.
| | | | | | | |
Collapse
|
33
|
Esposito C, Fasoli G, Plati AR, Bellotti N, Conte MM, Cornacchia F, Foschi A, Mazzullo T, Semeraro L, Dal Canton A. Long-term exposure to high glucose up-regulates VCAM-induced endothelial cell adhesiveness to PBMC. Kidney Int 2001; 59:1842-9. [PMID: 11318955 DOI: 10.1046/j.1523-1755.2001.0590051842.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND The changes induced on endothelial cells by a long-term exposure to high glucose, a situation that mimics the hyperglycemia of diabetics, have not yet been determined. We compared short- and long-term effects of elevated glucose on macrovascular and microvascular endothelial cells. METHODS Endothelial cells were grown in high-glucose media for 24 hours and for 8 weeks. Cell proliferation was evaluated by cell counting, apoptosis and expression of adhesion molecules by flow cytometry; nitric oxide (NO) by measuring the concentration of nitrite/nitrate in the cell supernatant; alpha 2(IV) collagen mRNA and protein by reverse transcriptase-polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. The adhesion of peripheral blood mononuclear cells (PBMCs) to endothelial cells was evaluated by adhesion assay. In some experiments, endothelial cells were preincubated with anti-vascular cell adhesion molecule-1 (VCAM-1) and anti-receptor for advanced glycation end product (RAGE) blocking antibodies. RESULTS At 24 hours, but not at 8 weeks, high glucose increased endothelial cell proliferation and apoptosis. High glucose did not modify NO synthesis at 24 hours and 8 weeks. Collagen production and expression were increased only after eight weeks. VCAM-1 but not intercellular adhesion molecule-1 was up-regulated after 8 weeks, a change not observed after 24 hours. The adhesion of PBMCs was significantly increased at eight weeks and was completely abrogated by anti--VCAM-1 and by anti-RAGE antibodies. After 24 hours, there was a modest increase of PBMC adhesion that was not blunted by anti-RAGE antibodies. CONCLUSIONS Increased adhesion of PBMCs, caused by up-regulation of VCAM-1 with a mechanism involving advanced glycation end product (AGE) adducts, and augmented collagen deposition are critical effects of long-term high glucose on endothelial cells, and may eventually promote the atherosclerotic process.
Collapse
Affiliation(s)
- C Esposito
- Unit of Nephrology, Dialysis and Transplantation, IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
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: 5.9] [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
|
35
|
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
|
36
|
Schofer J, Schlüter M, Rau T, Hammer F, Haag N, Mathey DG. Influence of treatment modality on angiographic outcome after coronary stenting in diabetic patients: a controlled study. J Am Coll Cardiol 2000; 35:1554-9. [PMID: 10807460 DOI: 10.1016/s0735-1097(00)00574-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVES This retrospective study was designed to determine the six-month angiographic outcome after stenting of native coronary arteries in insulin-treated (ITDM) and non-ITDM patients with diabetes mellitus (DM) and compare the results with those in non-DM patients. BACKGROUND The influence of the treatment modality for DM on restenosis in patients undergoing coronary artery stenting has not been elucidated sufficiently. METHODS A total of 1,439 (70%) of 2,061 patients underwent repeated angiography within six months of coronary stenting. The ITDM and non-ITDM (oral hypoglycemic drugs or diet) were documented in 48 (3.3%) and 177 patients (12.3%), respectively, leaving 1,214 non-DM patients. RESULTS Baseline reference vessel diameter tended to be smaller in ITDM patients (mean, 2.73 mm) than in non-DM and non-ITDM patients (2.88 mm and 2.85 mm, respectively). However, percent diameter stenosis was not different. The median number of stents deployed was 1; median stent length was 15 mm. Statistically significant differences were present after stenting for the means of minimal lumen diameter (MLD) and acute gain between ITDM patients (MLD: 2.67 mm, acute gain: 1.98 mm) and non-DM patients (MLD: 2.81 mm, acute gain: 2.16 mm). At follow-up, percent diameter stenosis, late lumen loss and loss index were significantly higher in both non-ITDM lesions (42%, 1.14 mm and 0.56, respectively) and ITDM lesions (48%, 1.26 mm and 0.65, respectively) than in non-DM lesions (35%, 0.96 mm and 0.45, respectively). The corresponding differences between non-ITDM and ITDM lesions did not reach statistical significance. Restenosis rates in non-DM, non-ITDM and ITDM lesions were 23.8%, 32.8% (p = 0.013 vs. non-DM) and 39.6% (p = 0.02 vs. non-DM, p = 0.477 vs. non-ITDM), respectively. CONCLUSIONS This study showed that compared with stenting in non-DM patients, stenting of native coronary arteries in DM patients is associated with significantly increased lumen renarrowing, regardless of the treatment modality for DM.
Collapse
Affiliation(s)
- J Schofer
- Center for Cardiology Othmarschen, Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
37
|
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
|
38
|
Ichiki T, Funakoshi Y, Ito K, Takeshita A. Expression of monocyte chemoattractant protein-1 by nonenzymatically glycated albumin (Amadori adducts) in vascular smooth muscle cells. Biochem Biophys Res Commun 2000; 269:666-70. [PMID: 10720473 DOI: 10.1006/bbrc.2000.2350] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The biological effects of Amadori adducts that are early nonenzymatically glycated protein on vascular cells were poorly defined. We examined the effect of glycated serum albumin (GA) on the expression of monocyte chemoattractant protein-1(MCP-1) that is an important chemokine recruiting monocyte to blood vessel. GA increased MCP-1 mRNA expression with a peak after 3 h of stimulation. The induction of MCP-1 by GA was dose-dependent. The MCP-1 mRNA expression by GA was completely inhibited by PD98059 and genistein that inhibit mitogen activated protein (MAP) kinase kinase and tyrosine kinase, respectively. N-Acetylcysteine, a potent antioxidant, also suppressed the GA-induced MCP-1 expression. These results suggest that GA induces production of reactive oxygen species and activates tyrosine kinase and MAP kinase in VSMC. Activation of these signals results in MCP-1 expression. GA-induced MCP-1 expression may be one of the mechanisms by which the diabetic patients suffer from accelerated atherosclerosis.
Collapse
Affiliation(s)
- T Ichiki
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | | | | | | |
Collapse
|
39
|
Takemoto M, Yokote K, Nishimura M, Shigematsu T, Hasegawa T, Kon S, Uede T, Matsumoto T, Saito Y, Mori S. Enhanced expression of osteopontin in human diabetic artery and analysis of its functional role in accelerated atherogenesis. Arterioscler Thromb Vasc Biol 2000; 20:624-8. [PMID: 10712383 DOI: 10.1161/01.atv.20.3.624] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have previously reported that high glucose stimulates osteopontin (OPN) expression through protein kinase C-dependent pathways as well as hexosamine pathways in cultured rat aortic smooth muscle cells. The finding prompted us to study in vivo expression of OPN in diabetes mellitus. In the present study, we found by immunohistochemistry that medial layers of the carotid arteries of streptozotocin-induced diabetic rats and the forearm arteries of diabetic patients stained positively for OPN antibodies, whereas the staining from arteries of control rats and nondiabetic patients was negative. We also found that OPN stimulated the migration and enhanced platelet-derived growth factor (PDGF)-mediated DNA synthesis of cultured rat aortic smooth muscle cells. OPN and PDGF synergistically activated focal adhesion kinase as well as extracellular signal-regulated kinase; this finding seems to explain the OPN-induced enhancement of PDGF-mediated DNA synthesis. Taken together, our present results raise a possibility that OPN plays a role in the development of diabetic vascular complications.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Aorta/cytology
- Aorta/enzymology
- Arteriosclerosis/etiology
- Arteriosclerosis/metabolism
- Becaplermin
- Carotid Arteries/metabolism
- Cell Movement/drug effects
- Cell Movement/physiology
- Cells, Cultured
- DNA/biosynthesis
- DNA-Binding Proteins
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 2/metabolism
- Diabetic Angiopathies/metabolism
- Enzyme Activation/drug effects
- Female
- Focal Adhesion Kinase 1
- Focal Adhesion Protein-Tyrosine Kinases
- Forearm/blood supply
- Humans
- Male
- Microcirculation/physiology
- Middle Aged
- Mitogen-Activated Protein Kinases/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/enzymology
- Osteopontin
- Platelet-Derived Growth Factor/pharmacology
- Protein-Tyrosine Kinases/metabolism
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-sis
- Rats
- Rats, Wistar
- Sialoglycoproteins/biosynthesis
- Sialoglycoproteins/pharmacology
- Transcription Factors
- ets-Domain Protein Elk-1
Collapse
Affiliation(s)
- M Takemoto
- Second Department of Internal Medicine, School of Medicine, Chiba University, Chiba, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kasuya Y, Nakamura J, Hamada Y, Nakayama M, Sasaki H, Komori T, Chaya S, Watanabe G, Naruse K, Nakashima E, Kato K, Hotta N. An aldose reductase inhibitor prevents the glucose-induced increase in PDGF-beta receptor in cultured rat aortic smooth muscle cells. Biochem Biophys Res Commun 1999; 261:853-8. [PMID: 10441515 DOI: 10.1006/bbrc.1999.1111] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To examine the role of platelet-derived growth factor (PDGF) and the polyol pathway in the growth activity of smooth muscle cells (SMCs), [(3)H]-thymidine incorporation, [(125)I]-PDGF-BB binding and expression of PDGF-beta receptor protein were measured in rat aortic SMCs cultured with 5.5 or 20 mM glucose with or without anti-PDGF antibody or an aldose reductase inhibitor, epalrestat. SMCs cultured with 20 mM glucose demonstrated an accelerated thymidine incorporation compared with SMCs cultured with 5.5 mM glucose, which was prevented by anti-PDGF antibody. This acceleration of growth activity by 20 mM glucose was accompanied by an increase in PDGF-BB binding, which was due to the increased number of PDGF-beta receptors and the overexpression of PDGF-beta receptor protein. Epalrestat prevented all these abnormalities. These observations suggest that polyol pathway hyperactivity plays an important role in the proliferation of SMCs which may be mediated through the accelerated expression of PDGF-beta receptor protein.
Collapse
Affiliation(s)
- Y Kasuya
- The Third Department of Internal Medicine, Nagoya University School of Medicine, Nagoya, 466-8550, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Avena R, Mitchell ME, Carmody B, Arora S, Neville RF, Sidaway AN. Insulin-like growth factor-1 receptors mediate infragenicular vascular smooth muscle cell proliferation in response to glucose and insulin not by insulin receptors. Am J Surg 1999; 178:156-61. [PMID: 10487270 DOI: 10.1016/s0002-9610(99)00150-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE Vascular smooth muscle cell (VSMC) proliferation is an early event in the pathogenesis of atherosclerosis. Insulin and glucose are known to stimulate the growth of VSMC. Cell membrane receptors play an important role in the proliferation of VSMC in response to growth factors. Insulin and insulin-like growth factor-1 (IGF-1) have demonstrated a cross reactivity for receptor binding and function. By using monoclonal antibodies directed against insulin (IRA) and IGF-1 (IGF-1RA) receptors, we attempt to further delineate the mechanism for the proliferation of VSMC in response to insulin and glucose. METHODS Human infragenicular VSMC isolated from diabetic patients undergoing below-knee amputations were used. Cells from passages 3 to 6 were grown in serum-free media with a glucose concentrations of 0.1% or 0.2%, both with and without insulin (100 ng/mL). The baseline cell density was 4,635 +/- 329 cells/mL. IRA or IGF-1RA was added to the media, with the control group receiving neither antibody. Cells were grown in 5% CO2 at 37 degrees C for 6 days. Analysis of variance was used for statistical analysis, with P <0.05 considered significant. In addition, DNA synthesis was measured using thymidine incorporation assays in the same groups of cells receiving IRA, IGF-1RA, and no antibody. RESULTS IGF-1RA prevented the proliferation of VSMC in response to insulin and glucose, while IRA had no effect on cell growth. There was no significant growth when IGF-1RA was added to the media, while the control group and the group receiving IRA demonstrated significant growth compared with the baseline concentration of 4,635 +/- 329 cells/mL at all concentrations of insulin and glucose. [3H]thymidine incorporation assays confirmed the cell count results. CONCLUSIONS These results suggest that the mitogenic effects of insulin and glucose on infragenicular VSMC are due to stimulation of the IGF-1 receptor. VSMC antiproliferative strategies employing receptor blockade should be directed against the IGF-1 receptor, not the insulin receptor.
Collapse
MESH Headings
- Analysis of Variance
- Antibodies, Monoclonal
- Arteriosclerosis/etiology
- Arteriosclerosis/pathology
- Cell Count/drug effects
- Cell Division/drug effects
- Cell Division/physiology
- Cells, Cultured
- Cross Reactions
- Culture Media, Serum-Free
- DNA/biosynthesis
- Diabetes Mellitus/pathology
- Glucose/administration & dosage
- Glucose/pharmacology
- Humans
- Insulin/administration & dosage
- Insulin/pharmacology
- Mitogens/administration & dosage
- Mitogens/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Radiopharmaceuticals
- Receptor, IGF Type 1/drug effects
- Receptor, IGF Type 1/physiology
- Receptor, Insulin/drug effects
- Receptor, Insulin/physiology
- Thymidine/metabolism
- Tritium
Collapse
Affiliation(s)
- R Avena
- Department of Surgery, Veterans Affairs Medical Center, Washington, DC, USA
| | | | | | | | | | | |
Collapse
|
42
|
Zhou YF, Yu ZX, Wanishsawad C, Shou M, Epstein SE. The immediate early gene products of human cytomegalovirus increase vascular smooth muscle cell migration, proliferation, and expression of PDGF beta-receptor. Biochem Biophys Res Commun 1999; 256:608-13. [PMID: 10080946 DOI: 10.1006/bbrc.1999.0387] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Evidence suggests that human cytomegalovirus (HCMV) infection contributes to the development of atherosclerosis and restenosis. Because smooth muscle cell (SMC) proliferation and migration are crucial events of both processes, and because PDGF beta-receptor modulates SMC migration, we determined whether HCMV infection affects SMC proliferation, migration, and PDGF beta-receptor expression. We employed a SMC model in which HCMV infection leads to expression of only the immediate early (IE) HCMV gene products-HCMV infection of rat SMCs. We found that HCMV infection significantly (i) increased SMC proliferation (from 0.9 x 10(6) +/- 0.024 x 10(6) to 1.4 x 10(6) +/- 0.051 x 10(6) cells/well, p < 0.001); (ii) augmented SMC migration toward PDGF (from 64 +/- 37 to 116 +/- 51 cells/high power field; p < 0.01); and (iii) enhanced PDGF beta-receptor expression in a time-dependent fashion. We conclude that HCMV infection of rat SMCs increases SMC proliferation, migration, and PDGF beta-receptor expression. These findings identify further mechanisms by which CMV may contribute to the development of atherosclerosis and restenosis.
Collapse
MESH Headings
- Animals
- Antigens, Viral/genetics
- Antigens, Viral/physiology
- Aorta
- Becaplermin
- Blotting, Western
- Cell Count/drug effects
- Cell Division/drug effects
- Cell Movement/drug effects
- Cells, Cultured
- Cytomegalovirus/genetics
- Cytomegalovirus/physiology
- Dose-Response Relationship, Drug
- Genes, Immediate-Early
- Humans
- Immediate-Early Proteins/genetics
- Immediate-Early Proteins/physiology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/virology
- Platelet-Derived Growth Factor/pharmacology
- Proto-Oncogene Proteins c-sis
- Rats
- Receptor, Platelet-Derived Growth Factor beta
- Receptors, Platelet-Derived Growth Factor/biosynthesis
- Time Factors
- Transcription, Genetic
Collapse
Affiliation(s)
- Y F Zhou
- Cardiovascular Research Foundation, Washington Hospital Center, Washington, DC, 20010, USA.
| | | | | | | | | |
Collapse
|
43
|
Fukumoto H, Naito Z, Asano G, Aramaki T. Immunohistochemical and morphometric evaluations of coronary atherosclerotic plaques associated with myocardial infarction and diabetes mellitus. J Atheroscler Thromb 1999; 5:29-35. [PMID: 10077455 DOI: 10.5551/jat1994.5.29] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Immunohistochemical and morphometrical studies were performed to elucidate the specificity of atherosclerosis in the descending branch (the segments 5 and 6) of the left coronary artery associated with acute myocardial infarction (AMI) in the anterior wall of the heart and non-insulin-dependent diabetes mellitus (NIDDM). The NIDDM without AMI group showed diffuse intimal thickening with smooth muscle cells, combined with much more intense immunostaining of tenascin than the non diabetic groups. The AMI without NIDDM group showed atheromatous thickening with decreased smooth muscle cells, a large number of macrophage and TUNEL-positive cells compared with the groups without AMI. However, the AMI with NIDDM group revealed atherosclerotic lesion with decreased smooth muscle cells, increased macrophages and TUNEL positive cells associated with the increased localization of tenascin and TGF-beta1 compared with the control. These findings suggest that the specificity of coronary atherosclerosis in diabetic patients may be the extensive atherosclerotic changes associated with increased tenascin. In AMI with NIDDM, increased TGF beta1 may induce apoptosis in the atheroma and coronary dysfunction, contributing to the development of acute myocardial infarction.
Collapse
Affiliation(s)
- H Fukumoto
- Department of Pathology, Nippon Medical School, Tokyo, Japan
| | | | | | | |
Collapse
|
44
|
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: 3.2] [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
|
45
|
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
|
46
|
Etienne P, Parés-Herbuté N, Mani-Ponset L, Gabrion J, Rabesandratana H, Herbuté S, Monnier L. Phenotype modulation in primary cultures of aortic smooth muscle cells from streptozotocin-diabetic rats. Differentiation 1998; 63:225-36. [PMID: 9745713 DOI: 10.1111/j.1432-0436.1998.00225.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Diabetes mellitus is a major risk factor for atherosclerosis. In atherosclerotic lesions, arterial smooth muscle cells (SMC) change from a contractile to a synthetic phenotype characterized by active proliferation. A similar phenotype modulation occurs in vitro when isolated arterial SMC are grown in culture and is characterized by both changes in cell morphology and a typical switch in actin isoform expression. In this study, we examined the influence of streptozotocin (STZ)-induced diabetes on the differentiation state and the phenotype modulation of cultured rat aortic SMC. We used transmission electron microscopy to study the fine structure of STZ-diabetic and non-diabetic SMC in primary culture and immunological methods for the determination of the proportions of alpha-smooth muscle actin (alpha-SM) and nonmuscle beta-actin (beta-NM) isoforms. Cultured STZ-diabetic SMC exhibited a large cytoplasmic volume, rich in rough endoplasmic reticulum, when compared with cultured non-diabetic SMC. alpha-SM, organized in stress fibers, was less homogeneously and abundantly distributed and by contrast, beta-NM was more abundant in STZ-diabetic than in non-diabetic SMC. Cytofluorimetric analyses demonstrated that the alpha-SM content was reduced in freshly STZ-diabetic SMC. Furthermore, during logarithmic growth of cultured SMC, the decrease of alpha-SM was more important in STZ-diabetic than in non-diabetic SMC. Immunoblotting of actin isoforms confirmed that expression of beta-NM was more important in STZ-diabetic than in non-diabetic SMC even in freshly isolated cells. The results suggest that SMC from STZ-diabetic rats express a more dedifferentiated state and undergo a more rapid phenotypic modulation in primary cultures than SMC from non-diabetic rats. Therefore, diabetes could induce changes in the phenotype of arterial SMC which might be associated with the onset or progression of the atherogenic process.
Collapse
MESH Headings
- Actins/analysis
- Actins/biosynthesis
- Animals
- Aorta, Thoracic/cytology
- Aorta, Thoracic/pathology
- Aorta, Thoracic/ultrastructure
- Cell Differentiation
- Cell Division
- Cells, Cultured
- Diabetes Mellitus, Experimental/pathology
- Flow Cytometry
- Kinetics
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/ultrastructure
- Phenotype
- Rats
- Rats, Wistar
- Reference Values
- Time Factors
Collapse
Affiliation(s)
- P Etienne
- Department of Metabolism, Lapeyronie Hospital, Montpellier, France
| | | | | | | | | | | | | |
Collapse
|
47
|
SILVA JOSEA, WHITE CHRISTOPHERJ. Diabetes Mellitus as a Risk Factor for Development of Vulnerable (Unstable) Coronary Plaque: A Review of Possible Mechanisms. J Interv Cardiol 1998. [DOI: 10.1111/j.1540-8183.1998.tb00091.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
48
|
Tazi KA, Trombino C, Moreau R, Lebrec D. Responsiveness to growth factors in aortic vascular smooth muscle cells from rats with cirrhosis. THE AMERICAN JOURNAL OF PHYSIOLOGY 1997; 273:G883-90. [PMID: 9357831 DOI: 10.1152/ajpgi.1997.273.4.g883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Hemodynamic changes in cirrhosis may be associated with alterations in aortic vascular smooth muscle cell (AVSMC) function. The present study compared the proliferative response to serum and growth factors in cirrhotic and control AVSMC. Serum from cirrhotic rats, cirrhotic cell lysates, and the conditioned medium of cultured cirrhotic AVSMC induced an increase in [3H]thymidine incorporation in control but not in cirrhotic AVSMC. Platelet-derived growth factor-beta (PDGF-BB) induced a greater increase in [3H]thymidine incorporation in cirrhotic than in control cells. [3H]thymidine incorporation induced by cirrhotic conditioned medium was blocked by anti-PDGF antibody. Immunoblot studies showed that the anti-PDGF antibody recognized a 30-kDa protein in the conditioned medium of cirrhotic AVSMC culture, a protein corresponding to PDGF. Binding studies of PDGF-BB indicated a twofold increase in receptor density in cirrhotic AVSMC with no alteration in affinity for PDGF-BB. We conclude that an increased responsiveness of cirrhotic AVSMC to the PDGF could contribute to alterations in AVSMC and muscle cell tone that may play a role in the hemodynamic changes in cirrhosis.
Collapse
MESH Headings
- Animals
- Antibodies/pharmacology
- Aorta, Thoracic/cytology
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/pathology
- Becaplermin
- Cell Division/drug effects
- Cell Survival
- Cells, Cultured
- Culture Media, Conditioned
- Fibroblast Growth Factor 2/pharmacology
- Kinetics
- Liver Cirrhosis, Experimental/blood
- Liver Cirrhosis, Experimental/pathology
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Platelet-Derived Growth Factor/immunology
- Platelet-Derived Growth Factor/pharmacology
- Platelet-Derived Growth Factor/physiology
- Proto-Oncogene Proteins c-sis
- Rats
- Rats, Sprague-Dawley
- Thymidine/metabolism
Collapse
Affiliation(s)
- K A Tazi
- Laboratoire d'Hémodynamique Splanchnique et de Biologie Vasculaire, Institut National de la Santé et de la Recherche Médicale, Hôpital Beaujon, Clichy, France
| | | | | | | |
Collapse
|
49
|
Yoo HJ, Kozaki K, Akishita M, Watanabe M, Eto M, Nagano K, Sudo N, Hashimoto M, Kim S, Yoshizumi M, Toba K, Ouchi Y. Augmented Ca2+ influx is involved in the mechanism of enhanced proliferation of cultured vascular smooth muscle cells from spontaneously diabetic Goto-Kakizaki rats. Atherosclerosis 1997; 131:167-75. [PMID: 9199269 DOI: 10.1016/s0021-9150(97)06098-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
To investigate whether augmented calcium influx is involved in the mechanism of the enhanced proliferation of vascular smooth muscle cells (VSMCs) in diabetes, we studied the association between proliferation and cytosolic free calcium concentration ([Ca2+]i) in cultured aortic VSMCs from spontaneously diabetic Goto-Kakizaki (GK) and Wistar rats. Serum, angiotensin II and Bay K 8644, a voltage-dependent Ca2+ channel (VDC) agonist, stimulated the proliferation of VSMCs; the magnitude was greater in VSMCs from GK than Wistar rats. VDC blockers, verapamil and nicardipine, inhibited Bay K 8644-induced cell proliferation, and the difference in the proliferation of VSMCs between GK and Wistar rats disappeared. Angiotensin II-induced proliferation was only partially inhibited by VDC blockers, and enhanced proliferation of GK-VSMCs was still observed. Bay K 8644 and angiotensin II increased [Ca2+]i, and the increase was augmented in GK-VSMCs. Bay K 8644-induced [Ca2+]i increase was completely inhibited by pretreatment with verapamil or removal of extracellular Ca2+, suggesting that VDC is associated with this increase. Although angiotensin II-induced [Ca2+]i increase was not affected by verapamil, removal of extracellular Ca2+ slightly but significantly attenuated angiotensin II-induced [Ca2+]i increase, suggesting that VDC blocker-insensitive receptor-activated Ca2+ influx is involved. These results indicate that augmented Ca2+ influx via VDC and a receptor-activated pathway may be involved in the mechanism of the enhanced proliferation of VSMCs from GK rats.
Collapse
MESH Headings
- 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology
- Angiotensin II/pharmacology
- Animals
- Aorta, Thoracic
- Blood Glucose/metabolism
- Calcium/metabolism
- Calcium Channel Agonists/pharmacology
- Calcium Channel Blockers/pharmacology
- Calcium Channels/drug effects
- Calcium Channels/metabolism
- Cell Count
- Cell Division/drug effects
- Cells, Cultured
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Insulin/blood
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Nicardipine/pharmacology
- Rats
- Rats, Wistar
- Vasoconstrictor Agents/pharmacology
- Verapamil/pharmacology
Collapse
Affiliation(s)
- H J Yoo
- Department of Geriatrics, Faculty of Medicine, University of Tokyo, Bunkyo-ku, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Nishida M, Miyagawa JI, Tokunaga K, Yamamoto K, Keno Y, Kobatake T, Yoshida S, Nakamura T, Odaka H, Ikeda H, Hanafusa T, Yamashita S, Kameda-Takemura K, Matsuzawa Y. Early morphologic changes of atherosclerosis induced by ventromedial hypothalamic lesion in the spontaneously diabetic Goto-Kakizaki rat. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 1997; 129:200-7. [PMID: 9016856 DOI: 10.1016/s0022-2143(97)90140-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
It is generally thought that typical atherosclerotic lesions do not develop in the rodent. The Goto-Kakizaki (GK) rat is a nonobese strain in which a spontaneous type of non-insulin-dependent diabetes mellitus develops without apparent macroangiopathy. In our previous study, making ventromedial hypothalamic (VMH) lesions in GK rats induced hyperphagia and a further deterioration in glucose metabolism. In the current study, male GK rats in which VMH lesions were made were examined for vascular changes, with special reference to atherosclerotic lesions. Marked hyperglycemia in GK rats with VMH lesions (hereafter referred to as VMH lesion rats) was revealed over an observation period (plasma glucose levels 16 weeks after the operation: VMH lesion GK rats, 19.3 +/- 2.0 mmol/L, vs sham-operated GK rats, 10.1 +/- 1.3 mmol/L; p < 0.0001). Light microscopic observation of the descending aorta in VMH lesion GK rats 16 weeks after the surgery revealed that the intimal thickening and the number of infiltrating cells into the intima were significantly increased as compared with sham-operated GK rats (17531 +/- 3747 microm2 vs 3072 +/- 1192 microm2, p < 0.0001; 15.6 +/- 3.1 per one transverse section vs 6.8 +/- 2.5 per one transverse section, p < 0.0005). Electron microscopic observations demonstrated an increased number of microvilli and lysosomes in endothelial cells, infiltration of macrophages and lymphocytes into the intima, and migration of medial smooth muscle cells into the intima that are considered to be early events in atherosclerosis. These morphologic changes could be induced by a deterioration in glucose metabolism. This rat may thus be useful for studying the process of the initiation of atherosclerosis in diabetes mellitus.
Collapse
Affiliation(s)
- M Nishida
- Second Department of Internal Medicine, Osaka University Medical School, Suita, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|