1
|
Zhang Y, Dai D, Geng S, Rong C, Zou R, Leng X, Xiang J, Liu J, Ding J. PCSK9 expression in fibrous cap possesses a marker for rupture in advanced plaque. Vasc Med 2024; 29:483-495. [PMID: 38860436 DOI: 10.1177/1358863x241252370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
BACKGROUND To date, PCSK9 inhibitors are well known for eliminating cardiac and cerebral artery ischemia events by lowering the serum lipid level. However, the pathophysiological value of in-plaque PCSK9 expression is still unclear. METHODS Advanced plaques removed by carotid endarterectomy were sectioned and stained to identify the PCSK9 expression pattern and its co-expression with rupture-relevant markers. To investigate the correlation of PCSK9 expression with regional blood shear flow, hemodynamic characteristics were analyzed using computational fluid dynamics, and representative parameters were compared between PCSK9 positive and negative staining plaques. To explore this phenomenon in vitro, human aortic vascular smooth muscle cells were used to overexpress and knock down PCSK9. The impacts of PCSK9 modulations on mechanical sensor activity were testified by western blot and immunofluorescence. Real-time polymerase chain reaction was used to evaluate the transcription levels of downstream rupture-prone effectors. RESULTS PCSK9 distribution in plaque preferred cap and shoulder regions, residing predominantly in smooth muscle actin-positive cells. Cap PCSK9 expression correlated with fibrous cap thickness negatively and co-expressed with MMP-9, both pointing to the direction of plaque rupture. A hemodynamic profile indicated a rupture-prone feature of cap PCSK9 expression. In vitro, overexpression and knockdown of PCSK9 in human aortic vascular smooth muscle cells has positive modulation on mechanical sensor Yes-associated protein 1 (YAP) activity and transcription levels of its downstream rupture-prone effectors. Serial section staining verified in situ colocalization among PCSK9, YAP, and downstream effectors. CONCLUSIONS Cap PCSK9 possesses a biomarker for rupture risk, and its modulation may lead to a novel biomechanical angle for plaque interventions.
Collapse
MESH Headings
- Humans
- Plaque, Atherosclerotic
- Proprotein Convertase 9/genetics
- Proprotein Convertase 9/metabolism
- Rupture, Spontaneous
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/enzymology
- Fibrosis
- Cells, Cultured
- Matrix Metalloproteinase 9/genetics
- Matrix Metalloproteinase 9/metabolism
- Male
- Endarterectomy, Carotid
- Carotid Arteries/pathology
- Carotid Arteries/surgery
- Carotid Arteries/enzymology
- Carotid Arteries/metabolism
- Aged
- Mechanotransduction, Cellular
- Female
- Regional Blood Flow
- Carotid Stenosis/pathology
- Carotid Stenosis/genetics
- Carotid Stenosis/surgery
- Carotid Stenosis/metabolism
- Carotid Stenosis/enzymology
- Carotid Artery Diseases/genetics
- Carotid Artery Diseases/enzymology
- Carotid Artery Diseases/pathology
- Carotid Artery Diseases/metabolism
- Carotid Artery Diseases/surgery
Collapse
Affiliation(s)
- Yingying Zhang
- Department of Neurology, Fudan University Zhongshan Hospital, Shanghai, China
| | - Dongwei Dai
- Department of Neurovascular Center, Naval Medical University Changhai Hospital, Shanghai, China
- Department of Neurosurgery, Fudan University Huadong Hospital, Shanghai, China
| | | | | | - Rong Zou
- ArteryFlow Technology Co., Ltd, Hangzhou, China
| | | | | | - Jianmin Liu
- Department of Neurovascular Center, Naval Medical University Changhai Hospital, Shanghai, China
| | - Jing Ding
- Department of Neurology, Fudan University Zhongshan Hospital, Shanghai, China
| |
Collapse
|
2
|
Sabry MM, Dawood AF, Rashed LA, Sayed SM, Hassan S, Younes SF. Relation between resistin, PPAR-γ, obesity and atherosclerosis in male albino rats. Arch Physiol Biochem 2020; 126:389-398. [PMID: 30612469 DOI: 10.1080/13813455.2018.1550094] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Obesity and atherosclerosis are inflammatory states involving variable metabolic signals. The adipokine resistin is implicated in adipose tissue dysfunction and is modulated by PPARγ. In this study, resistin and PPARγ role is investigated in the development of CVS disease. Forty-eight Adult male albino rats were divided into control, obesity and atherosclerotic groups; each group is divided into two subgroups; with and without PPARγ agonist administration for 8 weeks. To assess pathological changes; lipid profile, inflammatory mediator, serum resistin level and resistin expression in adipose tissue were measured. Aorta is histopathologically evaluated. It was found that resistin expression is significantly correlated with lipid profile and inflammatory status in obesity and atherosclerotic groups, and PPARγ agonist administration significantly improves inflammatory status and dyslipidemic profile across studied groups (p < .05). Aortic wall shows histopathological evidence of atherosclerosis in obesity group which is more evident in atherosclerotic group, and milder changes upon receiving PPARγ agonist.
Collapse
Affiliation(s)
- Maha M Sabry
- Physiology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Amal F Dawood
- Physiology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of Basic Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, KSA
| | - Laila A Rashed
- Biochemistry Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Seham M Sayed
- Physiology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Sherif Hassan
- Department of Medical Education, California University of Sciences and Medicine, School of Medicine (CalMed-SOM), Irvine, USA
- Anatomy Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Sheren F Younes
- Department of Basic Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, KSA
- Pathology Department, Faculty of Medicine, Menoufia University, Cairo, Egypt
| |
Collapse
|
3
|
Wu Y, Zhang Y, Dai L, Wang Q, Xue L, Su Z, Zhang C. An apoptotic body-biomimic liposome in situ upregulates anti-inflammatory macrophages for stabilization of atherosclerotic plaques. J Control Release 2019; 316:236-249. [DOI: 10.1016/j.jconrel.2019.10.043] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 10/19/2019] [Accepted: 10/22/2019] [Indexed: 01/31/2023]
|
4
|
Murakami-Nishida S, Matsumura T, Senokuchi T, Ishii N, Kinoshita H, Yamada S, Morita Y, Nishida S, Motoshima H, Kondo T, Komohara Y, Araki E. Pioglitazone suppresses macrophage proliferation in apolipoprotein-E deficient mice by activating PPARγ. Atherosclerosis 2019; 286:30-39. [PMID: 31096071 DOI: 10.1016/j.atherosclerosis.2019.04.229] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 04/05/2019] [Accepted: 04/30/2019] [Indexed: 01/18/2023]
Abstract
BACKGROUND AND AIMS Local macrophage proliferation is linked to enhanced atherosclerosis progression. Our previous study found that troglitazone, a thiazolidinedione (TZD), suppressed oxidized low-density lipoprotein (Ox-LDL)-induced macrophage proliferation. However, its effects and mechanisms are unclear. Therefore, we investigated the effects of pioglitazone, another TZD, on macrophage proliferation. METHODS Normal chow (NC)- or high-fat diet (HFD)-fed apolipoprotein E-deficient (Apoe-/-) mice were treated orally with pioglitazone (10 mg/kg/day) or vehicle (water) as a control. Mouse peritoneal macrophages were used in in vitro assays. RESULTS Atherosclerosis progression was suppressed in aortic sinuses of pioglitazone-treated Apoe-/- mice, which showed fewer proliferating macrophages in plaques. Pioglitazone suppressed Ox-LDL-induced macrophage proliferation in a dose-dependent manner. However, treatment with peroxisome proliferator-activated receptor-γ (PPARγ) siRNA ameliorated pioglitazone-induced suppression of macrophage proliferation. Low concentrations (less than 100 μmol/L) of pioglitazone, which can suppress macrophage proliferation, activated PPARγ in macrophages, but did not induce macrophage apoptosis. Pioglitazone treatment did not induce TUNEL-positive cells in atherosclerotic plaques of aortic sinuses in Apoe-/- mice. CONCLUSIONS Pioglitazone suppressed macrophage proliferation through PPARγ without inducing macrophage apoptosis. These findings imply that pioglitazone could prevent macrovascular complications in diabetic individuals.
Collapse
Affiliation(s)
- Saiko Murakami-Nishida
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takeshi Matsumura
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| | - Takafumi Senokuchi
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Norio Ishii
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroyuki Kinoshita
- Department of Diabetes and Endocrinology, National Hospital Organization, Kumamoto Medical Center, Kumamoto, Japan
| | - Sarie Yamada
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yutaro Morita
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shuhei Nishida
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroyuki Motoshima
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Tatsuya Kondo
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Eiichi Araki
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; Center for Metabolic Regulation of Healthy Aging (CMHA), Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
5
|
Gao H, Li H, Li W, Shen X, Di B. Pioglitazone Attenuates Atherosclerosis in Diabetic Mice by Inhibition of Receptor for Advanced Glycation End-Product (RAGE) Signaling. Med Sci Monit 2017; 23:6121-6131. [PMID: 29278639 PMCID: PMC5749137 DOI: 10.12659/msm.907401] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Background Peroxisome proliferator-activated receptor-γ (PPAR-γ) exhibits anti-inflammatory and anti-diabetic properties, and is protective against cardiovascular diseases. This study aimed to determine the effects of a PPAR-γ agonist pioglitazone on atherogenesis in an ApoE knockout mouse (ApoE−/−) diabetic mouse model and in a cultured vascular smooth muscle cells (VSMCs) model. Material/Methods Male ApoE−/− mice were rendered diabetic by 5 daily intraperitoneal injections of streptozotocin. Pioglitazone (20 mg/kg/d) or PPAR-γ inhibitor GW9662 (1 mg/kg/d) were administered for 12 weeks. At the end of treatment, mice were killed and the aortae were isolated. Oil Red O staining was used to evaluate atherosclerotic plaque area. H&E staining was used to evaluate the number of complicated plaques. Western blotting and immunohistochemistry were used to determine the expression of advanced glycation end-products (RAGE) and PPAR-γ. The effects of pioglitazone and GW9662 on RAGE and PPAR-γ expression were examined in cultured primary mouse VSMCs in hyperglycemic conditions. Results Administration of pioglitazone in diabetic ApoE−/− mice successfully reduced atherosclerotic plaque area and the number of complicated plaques. Moreover, pioglitazone inhibited RAGE and stimulated PPAR-γ protein expression in atherosclerotic plaques of diabetic ApoE−/− mice. In cultured VSMCs upon high-glucose challenge, pioglitazone downregulated RAGE mRNA and protein expression. Blockade of PPAR-γ activity by GW9662 remarkably attenuated the inhibitory actions of pioglitazone on atherogenesis, both in diabetic ApoE−/− mice and in cultured VSMCs, upon high-glucose challenge. Conclusions Pioglitazone has a therapeutic effect on atherosclerosis in diabetes, and inhibition of RAGE signaling plays a critical role in mediating the beneficial effects of pioglitazone.
Collapse
Affiliation(s)
- Hongli Gao
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China (mainland)
| | - Hongwei Li
- Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China (mainland).,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Beijing, China (mainland)
| | - Weiping Li
- Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China (mainland).,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Beijing, China (mainland)
| | - Xuhua Shen
- Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China (mainland)
| | - Beibing Di
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China (mainland)
| |
Collapse
|
6
|
Tian Y, Chen T, Wu Y, Yang L, Wang L, Fan X, Zhang W, Feng J, Yu H, Yang Y, Zhou J, Yuan Z, Wu Y. Pioglitazone stabilizes atherosclerotic plaque by regulating the Th17/Treg balance in AMPK-dependent mechanisms. Cardiovasc Diabetol 2017; 16:140. [PMID: 29084546 PMCID: PMC5663071 DOI: 10.1186/s12933-017-0623-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/18/2017] [Indexed: 11/23/2022] Open
Abstract
Background Pioglitazone (PIO), a thiazolidinediones drug, is a well-known anti-diabetic medicine, but its anti-atherosclerotic effects remain controversial. Thus it is important to investigate the effects of PIO on atherogenesis and the relevant mechanisms. Methods For in vitro studies, primary cultured or AMP-activated protein kinase (AMPK) inhibited splenocytes were treated with oxidized low density lipoprotein (ox-LDL) or ox-LDL plus PIO. Percentage of T helper 17 (Th17) and regulatory T (Treg) cells were determined by flow cytometry. Expression of AMPK, interleukin-17 (IL-17) and forkhead box P3 (FoxP3) were detected by Western blots. For in vivo studies, apolipoprotein E–deficient (apoE−/−) mice fed with western diet were treated with PIO or vehicle for 8 weeks respectively. Percentage of Th17 and Treg cells in spleen were measured by immunohistochemical analysis. The atherosclerotic lesions were analyzed using oil red O staining, and collagen types I and III in atherosclerotic lesions were stained by Sirius red. Expression of IL-17 and FoxP3 were determined by quantitative polymerase chain reaction. Results In cultured primary splenocytes, PIO dramatically inhibited Th17 and raised Treg. Intriguingly, pharmacological and genetic AMPK inhibitions abolished PIO-induced Treg elevation and Th17 inhibition. Moreover, PIO significantly induced AMPK phosphorylation, decreased IL-17+ and increased FoxP3+ cells in spleen of apoE−/− mice. Finally, PIO did not alter plaque area, but intriguingly, stabilized atherosclerotic plaque through collagen induction in apoE−/− mice. PIO treatment also improved Th17/Treg balance in atherosclerotic lesions. Conclusions PIO exhibits anti-atherosclerotic effects for stabilization of atherosclerotic plaque through regulating the Th17/Treg balance in an AMPK-dependent manner.
Collapse
Affiliation(s)
- Yuling Tian
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Tao Chen
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yan Wu
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Lin Yang
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Lijun Wang
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Xiaojuan Fan
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Wei Zhang
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Jiahao Feng
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Hang Yu
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yanjie Yang
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Juan Zhou
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Zuyi Yuan
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China.
| | - Yue Wu
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
7
|
Ungvari Z, Valcarcel-Ares MN, Tarantini S, Yabluchanskiy A, Fülöp GA, Kiss T, Csiszar A. Connective tissue growth factor (CTGF) in age-related vascular pathologies. GeroScience 2017; 39:491-498. [PMID: 28875415 DOI: 10.1007/s11357-017-9995-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 08/23/2017] [Indexed: 12/20/2022] Open
Abstract
Connective tissue growth factor (CTGF, also known as CCN2) is a matricellular protein expressed in the vascular wall, which regulates diverse cellular functions including cell adhesion, matrix production, structural remodeling, angiogenesis, and cell proliferation and differentiation. CTGF is principally regulated at the level of transcription and is induced by mechanical stresses and a number of cytokines and growth factors, including TGFβ. In this mini-review, the role of age-related dysregulation of CTGF signaling and its role in a range of macro- and microvascular pathologies, including pathogenesis of aorta aneurysms, atherogenesis, and diabetic retinopathy, are discussed. A potential role of CTGF and TGFβ in regulation and non-cell autonomous propagation of cellular senescence is also discussed.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Marta Noa Valcarcel-Ares
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Gábor A Fülöp
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamas Kiss
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Anna Csiszar
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA.
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary.
| |
Collapse
|
8
|
Silva AKSE, Gomes FODS, Santos Silva BD, Ribeiro EL, Oliveira AC, Araújo SMDR, de Lima IT, Oliveira AGV, Rudnicki M, Abdalla DS, Lima MDCAD, Pitta IDR, Peixoto CA. Chronic LPSF/GQ-02 treatment attenuates inflammation and atherosclerosis development in LDLr−/− mice. Eur J Pharmacol 2016; 791:622-631. [DOI: 10.1016/j.ejphar.2016.09.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/25/2016] [Accepted: 09/28/2016] [Indexed: 12/12/2022]
|
9
|
Liu XH, Zhang QY, Pan LL, Liu SY, Xu P, Luo XL, Zou SL, Xin H, Qu LF, Zhu YZ. NADPH oxidase 4 contributes to connective tissue growth factor expression through Smad3-dependent signaling pathway. Free Radic Biol Med 2016; 94:174-84. [PMID: 26945889 DOI: 10.1016/j.freeradbiomed.2016.02.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 02/21/2016] [Accepted: 02/28/2016] [Indexed: 01/28/2023]
Abstract
Transforming growth factor-β (TGF-β)/Smad signaling has been implicated in connective tissue growth factor (CTGF) expression in vascular smooth muscle cells (VSMC). Reactive oxygen species (ROS) are involved in activation of TGF-β/Smad signaling. However, detailed mechanisms underlying the process remain unclear. In present study, we demonstrated TGF-β1 strongly induced CTGF expression, Smad3 activation, NADPH oxidase 4 (Nox4) expression and increased ROS production in primary rat VSMC in vitro. NADPH oxidases inhibitor diphenylene iodonium (DPI) eliminated TGF-β1-induced CTGF expression and ROS generation. In addition, small-interfering RNA (siRNA) silencing of Smad3 or Nox4 significantly suppressed TGF-β1-mediated CTGF expression in VSMC. Furthermore, Nox4 silencing or inhibition eliminated TGF-β1-induced Smad3 activation and interaction between Nox4 and Smad3. In vivo studies further identified a positive correlation of Nox4 levels with Smad3 activation and CTGF expression in atherosclerotic arteries of patients and animal models. These data established that a novel mechanistic link of Nox4-dependent activation of Smad3 to increased TGF-β1-induced CTGF in the process of vascular remodeling, which suggested a new potential pathway for therapeutic interventions.
Collapse
Affiliation(s)
- Xin-Hua Liu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Qiu-Yan Zhang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Li-Long Pan
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Si-Yu Liu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Peng Xu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Xiao-Ling Luo
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Si-Li Zou
- Department of Vascular Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Hong Xin
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Le-Feng Qu
- Department of Vascular Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China.
| | - Yi-Zhun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China; School of Pharmacy, Macau University of Science and Technology, Macau.
| |
Collapse
|
10
|
Soares e Silva AK, de Oliveira Cipriano Torres D, dos Santos Gomes FO, dos Santos Silva B, Lima Ribeiro E, Costa Oliveira A, dos Santos LAM, de Lima MDCA, Pitta IDR, Peixoto CA. LPSF/GQ-02 inhibits the development of hepatic steatosis and inflammation in a mouse model of non-alcoholic fatty liver disease (NAFLD). PLoS One 2015; 10:e0123787. [PMID: 25875942 PMCID: PMC4397012 DOI: 10.1371/journal.pone.0123787] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 03/08/2015] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) defines a wide spectrum of liver diseases that extends from simple steatosis to non-alcoholic steatohepatitis. Although the pathogenesis of NAFLD remains undefined, it is recognized that insulin resistance is present in almost all patients who develop this disease. Thiazolidinediones (TZDs) act as an insulin sensitizer and have been used in the treatment of patients with type 2 diabetes and other insulin-resistant conditions, including NAFLD. Hence, therapy of NAFLD with insulin-sensitizing drugs should ideally improve the key hepatic histological changes, while also reducing cardiometabolic and cancer risks. Controversially, TZDs are associated with the development of cardiovascular events and liver problems. Therefore, there is a need for the development of new therapeutic strategies to improve liver function in patients with chronic liver diseases. The aim of the present study was to assess the therapeutic effects of LPSF/GQ-02 on the liver of LDLR-/- mice after a high-fat diet. Eighty male mice were divided into 4 groups and two different experiments: 1-received a standard diet; 2-fed with a high-fat diet (HFD); 3–HFD+pioglitazone; 4–HFD+LPSF/GQ-02. The experiments were conducted for 10 or 12 weeks and in the last two or four weeks respectively, the drugs were administered daily by gavage. The results obtained with an NAFLD murine model indicated that LPSF/GQ-02 was effective in improving the hepatic architecture, decreasing fat accumulation, reducing the amount of collagen, decreasing inflammation by reducing IL-6, iNOS, COX-2 and F4 / 80, and increasing the protein expression of IκBα, cytoplasmic NFκB-65, eNOS and IRS-1 in mice LDLR -/-. These results suggest a direct action by LPSF/GQ-02 on the factors that affect inflammation, insulin resistance and fat accumulation in the liver of these animals. Further studies are being conducted in our laboratory to investigate the possible mechanism of action of LPSF/GQ-02 on hepatic lipid metabolism.
Collapse
Affiliation(s)
| | | | | | - Bruna dos Santos Silva
- Laboratório de Ultraestrutura, Centro de Pesquisa Aggeu Magalhães (FIOCRUZ), Pernambuco, Recife, Brazil
| | - Edlene Lima Ribeiro
- Laboratório de Ultraestrutura, Centro de Pesquisa Aggeu Magalhães (FIOCRUZ), Pernambuco, Recife, Brazil
| | - Amanda Costa Oliveira
- Laboratório de Ultraestrutura, Centro de Pesquisa Aggeu Magalhães (FIOCRUZ), Pernambuco, Recife, Brazil
| | | | - Maria do Carmo Alves de Lima
- Laboratório de planejamento e síntese de fármacos, Universidade federal de Pernambuco, Pernambuco, Recife, Brazil
| | - Ivan da Rocha Pitta
- Laboratório de planejamento e síntese de fármacos, Universidade federal de Pernambuco, Pernambuco, Recife, Brazil
| | - Christina Alves Peixoto
- Laboratório de Ultraestrutura, Centro de Pesquisa Aggeu Magalhães (FIOCRUZ), Pernambuco, Recife, Brazil
| |
Collapse
|
11
|
Kang SW, Kim MS, Kim HS, Kim Y, Shin D, Park JHY, Kang YH. Celastrol attenuates adipokine resistin-associated matrix interaction and migration of vascular smooth muscle cells. J Cell Biochem 2013; 114:398-408. [PMID: 22949384 DOI: 10.1002/jcb.24374] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 08/22/2012] [Indexed: 11/06/2022]
Abstract
Obesity instigates various health problems such as atherosclerosis, diabetes, and cancer. Resistin, an adipose tissue-specific secretory adipokine, operates endocrine functions through increasing insulin resistance. Vascular smooth muscle cells (SMC) migrate into the subendothelial space and proliferate, thereby contributing to neointimal formation in atherosclerosis and restenosis. The aim of this study was to elucidate whether celastrol obtained from Tripterygium wilfordii Hook, inhibited human aortic SMC migration. Celastrol capable of antagonizing inflammatory responses attenuated the resistin secretion from THP-1-derived macrophages. The macrophage-conditioned media promoted SMC proliferation and MMP-2 production, which was dampened by 10-100 nM celastrol. Celastrol encumbered the SMC migration in response to 50 ng/ml resistin, concomitant with the inhibition of induction of connective tissue growth factor and collagen I/IV. In addition, celastrol disabled human aortic SMC exposed to resistin from migrating. The resistin-induced shedding of integrin β2/β3 expression was demoted by celastrol, thereby contributing to the inhibition of collagen matrix-SMC interaction. Next, resistin-induced Toll-like receptor-4 (TLR-4) expression was abrogated by celastrol, indicating that TLR-4 was the resistin signaling receptor that was blocked by celastrol. Collectively, these results demonstrate that anti-inflammatory celastrol blunted the macrophage secretion of the adipokine resistin, and suppressed the SMC migration by disturbing the interaction between SMC and intimal collagen matrix. Therefore, celastrol may inhibit atherogenic migration of vascular SMC upon resistin loading by intimal macrophages within atherosclerotic lesions.
Collapse
Affiliation(s)
- Sang-Wook Kang
- Department of Food and Nutrition, Hallym University, Chuncheon, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
12
|
Huang J, Huang H, Wu M, Li J, Xie H, Zhou H, Liao E, Peng Y. Connective tissue growth factor induces osteogenic differentiation of vascular smooth muscle cells through ERK signaling. Int J Mol Med 2013; 32:423-9. [PMID: 23722620 DOI: 10.3892/ijmm.2013.1398] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 04/11/2013] [Indexed: 11/06/2022] Open
Abstract
Connective tissue growth factor (CTGF) plays an important role in the pathogenesis of atherosclerosis by promoting vascular smooth muscle cell (VSMC) growth, migration, apoptosis, adhesion and the secretion of matrix components. The osteogenic differentiation of VSMCs is essential in the development of vascular calcification. However, the role of CTGF in the transdifferentiation and calcification of VSMCs is unclear. In the present study, we examined whether CTGF stimulates VSMC transdifferentiation. Primary VSMCs were obtained from mouse thoracic aortas by enzymatic digestion and identified by immunostaining for smooth muscle specific α-actin antibody (α-SMA). VSMC calcification was induced by the addition of CTGF to the osteogenic mediaum containing 5-10% FBS in the presence of 0.25 mM ascorbic acid and 10 mM β-glycerophosphate for 14 days. Calcified cells were determined by Alizarin Red S staining. Our results revealed that CTGF induced the expression of several bone markers, including alkaline phosphatase (ALP), osteocalcin (OC), osteoprotegerin (OPG) and core-binding factor subunit α1 (Cbfα1)/runt-related transcription factor 2 (Runx2), as well as calcification. However, the inhibition of extracellular signal-regulated kinase (ERK) activity using the ERK-specific inhibitor, PD98059, blocked the induction of these proteins and VSMC calcification. Based on these data, we conclude that CTGF stimulates the transdifferentiation of VSMCs into osteoblasts and that the ERK signaling pathway appears to play a critical role in this process.
Collapse
Affiliation(s)
- Juan Huang
- Institute of Endocrinology and Metabolism, The Second Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Rosa BLM, Vicenta LC, Victor GC, Dolores LC, Fernando C, Ricardo GH, Lina B, Tinahones FJ. Alterations of specific biomarkers of metabolic pathways in vascular tree from patients with Type 2 diabetes. Cardiovasc Diabetol 2012; 11:86. [PMID: 22828168 PMCID: PMC3461422 DOI: 10.1186/1475-2840-11-86] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 07/24/2012] [Indexed: 12/29/2022] Open
Abstract
The aims of this study were to check whether different biomarkers of inflammatory, apoptotic, immunological or lipid pathways had altered their expression in the occluded popliteal artery (OPA) compared with the internal mammary artery (IMA) and femoral vein (FV) and to examine whether glycemic control influenced the expression of these genes. The study included 20 patients with advanced atherosclerosis and type 2 diabetes mellitus, 15 of whom had peripheral arterial occlusive disease (PAOD), from whom samples of OPA and FV were collected. PAOD patients were classified based on their HbA1c as well (HbA1c ≤ 6.5) or poorly (HbA1c > 6.5) controlled patients. Controls for arteries without atherosclerosis comprised 5 IMA from patients with ischemic cardiomyopathy (ICM). mRNA, protein expression and histological studies were analyzed in IMA, OPA and FV. After analyzing 46 genes, OPA showed higher expression levels than IMA or FV for genes involved in thrombosis (F3), apoptosis (MMP2, MMP9, TIMP1 and TIM3), lipid metabolism (LRP1 and NDUFA), immune response (TLR2) and monocytes adhesion (CD83). Remarkably, MMP-9 expression was lower in OPA from well-controlled patients. In FV from diabetic patients with HbA1c ≤6.5, gene expression levels of BCL2, CDKN1A, COX2, NDUFA and SREBP2 were higher than in FV from those with HbA1c >6.5. The atherosclerotic process in OPA from diabetic patients was associated with high expression levels of inflammatory, lipid metabolism and apoptotic biomarkers. The degree of glycemic control was associated with gene expression markers of apoptosis, lipid metabolism and antioxidants in FV. However, the effect of glycemic control on pro-atherosclerotic gene expression was very low in arteries with established atherosclerosis.
Collapse
Affiliation(s)
- Bernal-Lopez M Rosa
- Biomedical Research Laboratory, Endocrinology Department, Hospital Virgen de la Victoria, Malaga, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Soares E Silva AK, de Oliveira Cipriano Torres D, Santos Rocha SW, dos Santos Gomes FO, dos Santos Silva B, Donato MAM, Raposo C, Santos ACO, de Lima MDCA, Galdino SL, da Rocha Pitta I, de Souza JRB, Peixoto CA. Effect of new thiazolidine derivatives LPSF/GQ-02 and LPSF/GQ-16 on atherosclerotic lesions in LDL receptor-deficient mice (LDLR(-/-)). Cardiovasc Pathol 2012; 22:81-90. [PMID: 22795892 DOI: 10.1016/j.carpath.2012.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 05/25/2012] [Accepted: 05/28/2012] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Atherosclerotic cardiovascular disease is a chronic inflammatory condition. Thiazolidinediones (TZDs) are used to enhance sensitivity to insulin and have demonstrated a protective effect over a variety of cardiovascular markers and risk factors. Controversially, the TZDs are associated with the development of heart failure. Thus, lines of research have invested in the search for new molecules in order to obtain more selective and less harmful treatment alternatives for the pathogenesis of atherosclerosis and its risk factors. METHODS Animals were fed a diet rich in fat for 10 weeks. In the last 2 weeks, animals received either pioglitazone, LPSF/GQ-02, or LPSF/GQ-16 daily through gavage. At the end of the treatment, blood was collected for biochemical analysis and the aortas were dissected for subsequent analyses. RESULTS No changes in the blood lipid profile were found following the use of the drugs in comparison to the control. However, the new thiazolidine derivatives were more efficient in improving insulin resistance in comparison to pioglitazone and the control group. Morphometric analyses revealed that neither pioglitazone nor LPSF/GQ16 led to satisfactory effects over atherosclerosis. However, LPSF/GQ-02 led to a reduction in area of the atherosclerotic lesions. Ultrastructural analyses revealed extensive degeneration of the endothelium and an increase in apoptotic cells in the subendothelial space following the use of pioglitazone and LPSF/GQ-16. However, LPSF/GQ-02 caused minimal cell alterations in the aortic endothelium. Regarding markers, endothelial nitric oxide synthase (eNOS) and matrix metalloproteinase 9 (MMP-9), LPSF/GQ-16, and pioglitazone exerted similar effects, increasing the expression of MMP-9, and had no effect on the expression of eNOS compared with the control group. On the other hand, LPSF/GQ-02 was effective in reducing the expression of MMP-9 and increased eNOS significantly. CONCLUSIONS The results suggest that the new thiazolidine derivative LPSF/GQ-02 is a promising candidate for the treatment of atherosclerosis.
Collapse
MESH Headings
- Animals
- Aorta/drug effects
- Aorta/metabolism
- Aorta/ultrastructure
- Aortic Diseases/drug therapy
- Aortic Diseases/genetics
- Aortic Diseases/pathology
- Apoptosis/drug effects
- Atherosclerosis/blood
- Atherosclerosis/drug therapy
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Blood Glucose/drug effects
- Blood Glucose/metabolism
- Blotting, Western
- Cardiovascular Agents/pharmacology
- Cardiovascular Agents/toxicity
- Disease Models, Animal
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Immunohistochemistry
- Insulin/blood
- Insulin Resistance
- Lipids/blood
- Matrix Metalloproteinase 9/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Electron, Transmission
- Nitric Oxide Synthase Type III/metabolism
- Pioglitazone
- Plaque, Atherosclerotic
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Thiazolidinediones/pharmacology
- Thiazolidinediones/toxicity
- Thiazolidines/pharmacology
- Thiazolidines/toxicity
- Time Factors
Collapse
|
15
|
Ma Y, Wang W, Zhang J, Lu Y, Wu W, Yan H, Wang Y. Hyperlipidemia and atherosclerotic lesion development in Ldlr-deficient mice on a long-term high-fat diet. PLoS One 2012; 7:e35835. [PMID: 22558236 PMCID: PMC3338468 DOI: 10.1371/journal.pone.0035835] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 03/23/2012] [Indexed: 11/19/2022] Open
Abstract
Background Mice deficient in the LDL receptor (Ldlr−/− mice) have been widely used as a model to mimic human atherosclerosis. However, the time-course of atherosclerotic lesion development and distribution of lesions at specific time-points are yet to be established. The current study sought to determine the progression and distribution of lesions in Ldlr−/− mice. Methodology/Principal Findings Ldlr-deficient mice fed regular chow or a high-fat (HF) diet for 0.5 to 12 months were analyzed for atherosclerotic lesions with en face and cross-sectional imaging. Mice displayed significant individual differences in lesion development when fed a chow diet, whereas those on a HF diet developed lesions in a time-dependent and site-selective manner. Specifically, mice subjected to the HF diet showed slight atherosclerotic lesions distributed exclusively in the aortic roots or innominate artery before 3 months. Lesions extended to the thoracic aorta at 6 months and abdominal aorta at 9 months. Cross-sectional analysis revealed the presence of advanced lesions in the aortic sinus after 3 months in the group on the HF diet and in the innominate artery at 6 to 9 months. The HF diet additionally resulted in increased total cholesterol, LDL, glucose, and HBA1c levels, along with the complication of obesity. Conclusions/Significance Ldlr-deficient mice on the HF diet tend to develop site-selective and size-specific atherosclerotic lesions over time. The current study should provide information on diet induction or drug intervention times and facilitate estimation of the appropriate locations of atherosclerotic lesions in Ldlr−/− mice.
Collapse
Affiliation(s)
- Yanling Ma
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wenyi Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jie Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Youli Lu
- Central Laboratory, Shanghai Xuhui Central Hospital, Shanghai, China
| | - Wenyu Wu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hong Yan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yiping Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- * E-mail:
| |
Collapse
|
16
|
Thrombin induced connective tissue growth factor expression in rat vascular smooth muscle cells via the PAR-1/JNK/AP-1 pathway. Acta Pharmacol Sin 2012; 33:49-56. [PMID: 22212430 DOI: 10.1038/aps.2011.178] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
AIM To investigate the signaling pathways involved in thrombin-induced connective tissue growth factor (CTGF) expression in rat vascular smooth muscle cells (VSMCs). METHODS Experiments were preformed on primary rat aortic smooth muscle cells (RASMCs) and a rat VSMC line (A10). CTGF protein levels were measured using Western blotting. Luciferase reporter genes and dominant negative mutants (DNs) were used to investigate the signaling pathways mediating the induction of CTGF expression by thrombin. RESULTS Thrombin (0.3-3.0 U/mL) caused a concentration- and time-dependent increase in CTGF expression in both RASMCs and A10 cells. Pretreating A10 cells with the protease-activated receptor 1 (PAR-1) antagonist SCH79797 (0.1 μmol/L) significantly blocked thrombin-induced CTGF expression, while the PAR-4 antagonist tcY-NH(2) (30 μmol/L) had no effect. The PAR-1 agonist SFLLRN-NH(2) (300 μmol/L) induced CTGF expression, while the PAR-4 agonist GYPGQV-NH(2) (300 μmol/L) had no effect. Thrombin (1 U/mL) caused time-dependent phosphorylation of c-Jun N-terminal kinase (JNK). Pretreating with the JNK inhibitor SP600125 (3-30 μmol/L) or transfection with DNs of JNK1/2 significantly attenuated thrombin-induced CTGF expression. Thrombin (0.3-3.0 U/mL) increased activator protein-1 (AP-1)-luciferase activity, which was inhibited by the JNK inhibitor SP600125. The AP-1 inhibitor curcumin (1-10 μmol/L) concentration-dependently attenuated thrombin-induced CTGF expression. CONCLUSION Thrombin acts on PAR-1 to activate the JNK signaling pathway, which in turn initiates AP-1 activation and ultimately induces CTGF expression in VSMCs.
Collapse
|
17
|
Kang SW, Kim JL, Kwon GT, Lee YJ, Park JHY, Lim SS, Kang YH. Sensitive Fern (Onoclea sensibilis) Extract Suppresses Proliferation and Migration of Vascular Smooth Muscle Cells Inflamed by Neighboring Macrophages. Biol Pharm Bull 2011; 34:1717-23. [DOI: 10.1248/bpb.34.1717] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | - Jung-Lye Kim
- Department of Food and Nutrition, Hallym University
| | | | | | | | | | | |
Collapse
|
18
|
Tian Y, Yuan Z, Liu Y, Liu W, Zhang W, Xue J, Shen Y, Liang X, Chen T, Kishimoto C. Pioglitazone modulates the balance of effector and regulatory T cells in apolipoprotein E deficient mice. Nutr Metab Cardiovasc Dis 2011; 21:25-32. [PMID: 19819680 DOI: 10.1016/j.numecd.2009.07.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2009] [Revised: 06/29/2009] [Accepted: 07/22/2009] [Indexed: 11/18/2022]
Abstract
BACKGROUND AND AIMS Pioglitazone (PIO) affects T cell-mediated immunity through actions of peroxisome proliferator activated receptor γ (PPARγ). Effector and regulatory T cells control the development of atherosclerosis, a chronic inflammatory disease affecting the arterial blood vessels. The aim of this study was to examine whether PIO ameliorates atherosclerosis by altering the balance of effector and regulatory T cells. METHODS AND RESULTS To explore the effect of PIO on early and advanced atherosclerosis, apolipoprotein E deficient (ApoE-/-) mice were fed western diet and received PIO (20 mg/kg/day) by gastric gavage at 6 or 14 weeks of age, respectively for 8 weeks. Data showed PIO markedly inhibited early fatty streak formation. Further, although the advanced fibrofatty plaque sizes were not significantly reduced, the numbers of smooth muscle cells within lesions were increased and higher collagen concentrations were produced. In general, macrophage expression in lesions was decreased. Additionally, the expression of Foxp3(+) cells was increased in lesions and spleens in mice at all PIO treatment stages, whereas the CD4(+)IFN-γ(+)/CD4(+)IL-4(+) cell ratios were reduced. CONCLUSION PIO inhibited early atherosclerotic lesion formation and increased the stability of advanced atherosclerotic plaques in ApoE-/- mice, which was associated with altering the balance of effector and regulatory T cells.
Collapse
Affiliation(s)
- Y Tian
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Sharma AK, Srinivasan BP. Triple verses glimepiride plus metformin therapy on cardiovascular risk biomarkers and diabetic cardiomyopathy in insulin resistance type 2 diabetes mellitus rats. Eur J Pharm Sci 2009; 38:433-44. [PMID: 19765654 DOI: 10.1016/j.ejps.2009.09.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 08/23/2009] [Accepted: 09/06/2009] [Indexed: 12/17/2022]
Abstract
IRtype2DM patients are often treated with a combination of antidiabetic agents. Drugs with different complementary mechanisms of action frequently used in daily clinical practice but glycemic control with monotherapeutic attempts fail in the long run. To date, biomarkers for cardiovascular risk and insulin sensitivity with combination of triple oral hypoglycemic therapies are not fully revelled in view of additional cardiovascular risk reduction. In the present study, IRtype2DM induced by administering streptozotocin (90 mg/kg, i.p.) in neonatal rat model. IRtype2DM rats were selected by determining FPI [>60 pmol/l]; HOMA-IR & Hyperinsulinemic euglycemic clamp technique at 6 weeks and then treated for 8 weeks with (i) Metformin (120 mg/kg, o.d.)+Glimepiride (1mg/kg, o.d.), (ii) Metformin (265 mg/kg, o.d.)+Rosiglitazone (1mg/kg, o.d.)+Glimepiride (0.7 mg/kg, o.d.). At the end cardiovascular risk parameters evaluated by ELISA kits and insulin sensitivity were determined by HOMA-IR. In conclusion, triple oral hypoglycemic therapy improves glycemic control, insulin sensitivity, retards diabetic cardiomyopathy and does not increased body weight; decrease more detrimental inflammatory markers, increase interleukin-10 and adiponectin in neonatal streptozotocin-induced IRtype2DM Wistar Albino Rats. Triple therapy showed a synergistic effect and was promising in insulin resistance, better in additional cardiovascular risk reduction and those nonresponders to metformin add on glimepiride therapy.
Collapse
Affiliation(s)
- Ashish Kumar Sharma
- Department of Pharmacology, Suresh Gyan Vihar University, Mahal, Jagatpura, Jaipur, Rajasthan, Pincode 302025, India.
| | | |
Collapse
|
20
|
Harashima K, Hayashi J, Miwa T, Tsunoda T. Long-term pioglitazone therapy improves arterial stiffness in patients with type 2 diabetes mellitus. Metabolism 2009; 58:739-45. [PMID: 19446110 DOI: 10.1016/j.metabol.2008.09.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Accepted: 09/18/2008] [Indexed: 11/18/2022]
Abstract
Pioglitazone, a peroxisome proliferator-activated receptor gamma agonist, not only improves insulin resistance and glycemic control, but may also have additional beneficial vascular effects in patients with type 2 diabetes mellitus. We investigated whether pioglitazone had an influence on arterial stiffness, which is an independent predictor of cardiovascular events, in 204 patients with type 2 diabetes mellitus. A prospective, nonrandomized, open-label trial was performed that involved 41 patients treated with pioglitazone, 46 patients receiving sulfonylureas, 67 patients on insulin, and 50 patients on diet/exercise only. The follow-up period was 56 +/- 3 months. Arterial stiffness was evaluated by using the arterial stiffness index (ASI), which was based on analysis of the pulse wave amplitude pattern obtained during automated blood pressure measurement in the upper limb. The 4 groups had a similar baseline ASI, which was greater than the reference range in each group. Although antidiabetic therapies improved hemoglobin A(1c) and low-density lipoprotein cholesterol, ASI only decreased significantly in the pioglitazone group. Thus, pioglitazone improved abnormal arterial stiffness in patients with type 2 diabetes mellitus via a mechanism beyond the metabolic improvement. These findings may have important clinical implications in the use of pioglitazone in patients with type 2 diabetes mellitus.
Collapse
|
21
|
Nakaya H, Summers BD, Nicholson AC, Gotto AM, Hajjar DP, Han J. Atherosclerosis in LDLR-knockout mice is inhibited, but not reversed, by the PPARgamma ligand pioglitazone. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:2007-14. [PMID: 19435790 DOI: 10.2353/ajpath.2009.080611] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Thiazolidinediones, a class of drugs for the treatment of type-2 diabetes, are synthetic ligands for peroxisome proliferator-activated receptor-gamma. They have been demonstrated to possess cardioprotective effects in humans and anti-atherogenic properties in animal models. However, the question remains whether a peroxisome proliferator-activated receptor-gamma ligand can reverse the development of atherosclerosis. In this study, we tested the effects of pioglitazone on the development of established atherosclerosis in low-density lipoprotein receptor-null mice. We observed that atherosclerosis in low-density lipoprotein receptor-null mice progressed when mice were fed a high-fat diet. Pioglitazone treatment of atherogenic mice prevented this progression of atherosclerosis from its middle stages of disease, but was not able to reverse it. Withdrawal of the high-fat diet from mice with advanced atherosclerosis did not result in a reduction in lesion sizes. Pioglitazone treatment also had no effect on advanced atherosclerosis. Levels of high density lipoprotein cholesterol correlated inversely with lesion development when pioglitazone was given during lesion progression. However, pioglitazone had no effect on circulating high density lipoprotein levels in mice in which treatment was initiated following 14 weeks on the high-fat diet. These findings have implications for the analysis of therapeutic agents in murine models of atherosclerosis and the use of pioglitazone in patients with established atherosclerosis.
Collapse
Affiliation(s)
- Hideaki Nakaya
- Center of Vascular Biology, Department of Pathology, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | | | | | | | | | | |
Collapse
|
22
|
Zheng M, Ye S, Zhai Z, Chen Y, Li X, Yang G, Fan A, Wang Y. Rosiglitazone protects diabetic rats against kidney disease through the suppression of renal moncyte chemoattractant protein-1 expression. J Diabetes Complications 2009; 23:124-9. [PMID: 18413206 DOI: 10.1016/j.jdiacomp.2007.11.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2007] [Revised: 09/04/2007] [Accepted: 11/09/2007] [Indexed: 11/15/2022]
Abstract
Although the pathogenetic mechanisms of diabetic nephropathy (DN) have not been elucidated thoroughly, an inflammatory mechanism has been suggested to contribute to its development and progression. Moncyte chemoattractant protein (MCP)-1 is a chemokine that can attract macrophages and T cells from the circulation to the local kidney, then activate them, and ultimately injure the renal tissue. Recent studies have demonstrated that thiazolidinediones decrease urinary albumin (ALB) excretion, which may be partly related to its anti-inflammatory action. Therefore, the effects of rosiglitazone on renal inflammation and renal injury were investigated in streptozotocin (STZ)-induced diabetic rats in this study. We examined the urinary excretion rates of ALB, retinal-binding protein (RBP), and MCP-1 of normal control group (Group C, n=8), STZ-induced diabetes mellitus group (Group D, n=8), and diabetes plus rosiglitazone (5 mg x kg-1 x day-1) treatment group (Group R, n=8) at the eighth week. The renal tissues of diabetic rats were obtained for reverse transcriptase-polymerase chain reaction to examine the expression of MCP-1 mRNA. Our results showed that compared to normal control, urinary excretion rates of ALB, RBP, and MCP-1 were significantly increased in untreated diabetic rats at the eighth week. However, rosiglitazone treatment could markedly decrease all the parameters above. In addition, urinary excretion rate of MCP-1 showed positive correlations with urinary ALB excretion, urinary RBP excretion, and kidney/body weight. The expressions of MCP-1 mRNA in renal tissues were markedly up-regulated in untreated diabetic rats, and these could be notably reduced by rosiglitazone treatment. In conclusion, rosiglitazone may have a potential therapeutic target in DN, which may be partly attributed to lowering of the expression of MCP-1 in the local kidney and the urinary excretion of MCP-1.
Collapse
Affiliation(s)
- Mao Zheng
- Department of Endocrinology, Anhui Provincial Hospital affiliated to Anhui Medical University, Hefei, Anhui Province of China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Jaffa AA, Usinger WR, McHenry MB, Jaffa MA, Lipstiz SR, Lackland D, Lopes-Virella M, Luttrell LM, Wilson PWF. Connective tissue growth factor and susceptibility to renal and vascular disease risk in type 1 diabetes. J Clin Endocrinol Metab 2008; 93:1893-900. [PMID: 18319310 PMCID: PMC2386274 DOI: 10.1210/jc.2007-2544] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
OBJECTIVE We explored the relevance and significance of connective tissue growth factor (CTGF) as a determinant of renal and vascular complications among type 1 diabetic patients. METHODS AND RESULTS We measured the circulating and urinary levels of CTGF and CTGF N fragment in 1050 subjects with type 1 diabetes from the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) Study cohort. We found that hypertensive diabetic subjects have significantly higher levels of plasma log CTGF N fragment relative to normotensive subjects (P = 0.0005). Multiple regression analysis showed a positive and independent association between CTGF N fragment levels and log albumin excretion rate (P < 0.0001). In categorical analysis, patients with macroalbuminuria had higher levels of CTGF N fragment than diabetic subjects with or without microalbuminuria (P < 0.0001). Univariate and multiple regression analyses demonstrated an independent and significant association of log CTGF N fragment with the common and internal carotid intima-media thickness. The relative risk for increased carotid intima-media thickness was higher in patients with concomitantly elevated plasma CTGF N fragment and macroalbuminuria relative to patients with normal plasma CTGF N fragment and normal albuminuria (relative risk = 4.76; 95% confidence interval, 2.21-10.25; P < 0.0001). CONCLUSION These findings demonstrate that plasma CTGF is a risk marker of diabetic renal and vascular disease.
Collapse
Affiliation(s)
- Ayad A Jaffa
- Department of Medicine, Endocrinology-Diabetes-Medical Genetics, Medical University of South Carolina, 114 Doughty Street, Charleston, SC 29425, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Balakumar P, Kaur T, Singh M. Potential target sites to modulate vascular endothelial dysfunction: Current perspectives and future directions. Toxicology 2008; 245:49-64. [DOI: 10.1016/j.tox.2007.12.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2007] [Revised: 12/13/2007] [Accepted: 12/13/2007] [Indexed: 12/26/2022]
|
25
|
Abstract
In the last few years, there has been increasing focus on the impact of interventions on cardiovascular outcomes in patients with type 2 diabetes. Insulin resistance and hyperglycaemia often co-exist with a cluster of risk factors for coronary artery disease, but the underlying mechanisms leading to the development of such vascular complications are complex. The over-production of free radicals in patients suffering from diabetes results in a state of oxidative stress, which leads to endothelial dysfunction and a greater risk of atherosclerosis. Moreover, inflammatory factors which play a critical role in atherothrombosis and plaque rupture are often found to be at elevated levels in this patient population. Thiazolidinediones (TZDs) are now routinely used to manage glucose levels, and have been suggested to influence other cardiovascular risk factors and therefore the pathways leading to macrovascular events. Consequently, recent studies have investigated the anti-inflammatory and anti-atherogenic properties of TZDs. The data available up to the present time, in the context of the emerging cardiovascular outcome profiles of rosiglitazone and pioglitazone, will be discussed here.
Collapse
Affiliation(s)
- Antonio Ceriello
- Warwick Medical School, Clinical Sciences Research Institute, University Hospital, Coventry, Warwickshire, UK.
| |
Collapse
|
26
|
Zhou R, Bruns CM, Bird IM, Kemnitz JW, Goodfriend TL, Dumesic DA, Abbott DH. Pioglitazone improves insulin action and normalizes menstrual cycles in a majority of prenatally androgenized female rhesus monkeys. Reprod Toxicol 2007; 23:438-48. [PMID: 17306503 PMCID: PMC2705750 DOI: 10.1016/j.reprotox.2006.12.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Revised: 12/13/2006] [Accepted: 12/21/2006] [Indexed: 12/31/2022]
Abstract
PURPOSE OF THE STUDY To determine whether pioglitazone will improve menstrual cyclicity in a fetal programming model for polycystic ovary syndrome. BASIC PROCEDURES Eight prenatally androgenized (PA) and 5 control female rhesus monkeys of similar age, body weight and body mass index received an oral placebo daily for 6-7 months followed, after at least 90 days, by daily oral dosing with pioglitazone (3mg/kg) for an additional 6-7 months. Blood was sampled thrice weekly to monitor ovulatory function, and a variety of endocrine challenges were performed to quantify changes in ovarian, gonadotropin and glucoregulatory function. MOST IMPORTANT FINDINGS Pioglitazone normalized menstrual cycles in 5 out of 8 (62%) PA females (pioglitazone responsive; Pio(RESP)). Pioglitazone increased serum 17alpha-hydroxyprogesterone responses to an hCG injection in Pio(RESP) PA females, while diminishing serum progesterone, and increasing DHEA and estradiol responses to hCG in Pio(RESP) PA and all normal females. PRINCIPAL CONCLUSIONS Insulin resistance plays a mechanistic role in maintaining anovulation in a majority of PA female monkeys.
Collapse
Affiliation(s)
- Rao Zhou
- National Primate Research Center, University of Wisconsin, Madison, WI 53715, United States
| | | | | | | | | | | | | |
Collapse
|