1
|
Spadafora L, Quarta R, Martino G, Romano L, Greco F, Curcio A, Gori T, Spaccarotella C, Indolfi C, Polimeni A. From Mechanisms to Management: Tackling In-Stent Restenosis in the Drug-Eluting Stent Era. Curr Cardiol Rep 2025; 27:53. [PMID: 39932602 DOI: 10.1007/s11886-025-02193-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/06/2025] [Indexed: 02/14/2025]
Abstract
PURPOSE OF REVIEW Drug-eluting stent (DES) technology has greatly enhanced the safety and effectiveness of percutaneous coronary interventions (PCI). The aim of the present paper is to provide a comprehensive review of in-stent restenosis (ISR), focusing on the contemporary DES era, including its incidence, mechanisms, and imaging characterization. RECENT FINDINGS Despite the widespread use of DES and numerous improvements, recent clinical data indicate that ISR still occurs in 5-10% of PCI procedures, posing a considerable public health issue. The incidence, morphology, and clinical implications of ISR are determined by a complex interplay of several factors: the patient, stent, procedure, and vessel and lesion-related factors. Advancements in intracoronary imaging have provided greater insight into its patterns and underlying causes. Over time, treatment strategies have evolved, and current guidelines recommend an individualized approach using intracoronary imaging to characterize ISR's underlying substrate.
Collapse
Affiliation(s)
- Luigi Spadafora
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.
| | - Rossella Quarta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
- Division of Cardiology, Annunziata Hospital, 87100, Cosenza, Italy
| | - Giovanni Martino
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100, Catanzaro, Italy
| | - Letizia Romano
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100, Catanzaro, Italy
- Division of Cardiology, Annunziata Hospital, 87100, Cosenza, Italy
| | - Francesco Greco
- Division of Interventional Cardiology, Annunziata Hospital, 87100, Cosenza, Italy
| | - Antonio Curcio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
- Division of Cardiology, Annunziata Hospital, 87100, Cosenza, Italy
| | - Tommaso Gori
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100, Catanzaro, Italy
- Zentrum Für Kardiologie, Kardiologie I, University Medical Center Mainz and DZHK Standort Rhein-Main, Mainz, Germany
| | - Carmen Spaccarotella
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples Federico II, 80126, Naples, Italy
| | - Ciro Indolfi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Alberto Polimeni
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
- Division of Interventional Cardiology, Annunziata Hospital, 87100, Cosenza, Italy
| |
Collapse
|
2
|
Cao G, Xuan X, Zhang R, Hu J, Dong H. Gene Therapy for Cardiovascular Disease: Basic Research and Clinical Prospects. Front Cardiovasc Med 2021; 8:760140. [PMID: 34805315 PMCID: PMC8602679 DOI: 10.3389/fcvm.2021.760140] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/11/2021] [Indexed: 12/16/2022] Open
Abstract
In recent years, the vital role of genetic factors in human diseases have been widely recognized by scholars with the deepening of life science research, accompanied by the rapid development of gene-editing technology. In early years, scientists used homologous recombination technology to establish gene knock-out and gene knock-in animal models, and then appeared the second-generation gene-editing technology zinc-finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs) that relied on nucleic acid binding proteins and endonucleases and the third-generation gene-editing technology that functioned through protein-nucleic acids complexes-CRISPR/Cas9 system. This holds another promise for refractory diseases and genetic diseases. Cardiovascular disease (CVD) has always been the focus of clinical and basic research because of its high incidence and high disability rate, which seriously affects the long-term survival and quality of life of patients. Because some inherited cardiovascular diseases do not respond well to drug and surgical treatment, researchers are trying to use rapidly developing genetic techniques to develop initial attempts. However, significant obstacles to clinical application of gene therapy still exists, such as insufficient understanding of the nature of cardiovascular disease, limitations of genetic technology, or ethical concerns. This review mainly introduces the types and mechanisms of gene-editing techniques, ethical concerns of gene therapy, the application of gene therapy in atherosclerosis and inheritable cardiovascular diseases, in-stent restenosis, and delivering systems.
Collapse
Affiliation(s)
- Genmao Cao
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xuezhen Xuan
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Ruijing Zhang
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jie Hu
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Honglin Dong
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
3
|
An Autocrine Negative Feedback Loop Inhibits Dictyostelium discoideum Proliferation through Pathways Including IP3/Ca 2. mBio 2021; 12:e0134721. [PMID: 34154396 PMCID: PMC8262924 DOI: 10.1128/mbio.01347-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Little is known about how eukaryotic cells can sense their number or spatial density and stop proliferating when the local density reaches a set value. We previously found that Dictyostelium discoideum accumulates extracellular polyphosphate to inhibit its proliferation, and this requires the G protein-coupled receptor GrlD and the small GTPase RasC. Here, we show that cells lacking the G protein component Gβ, the Ras guanine nucleotide exchange factor GefA, phosphatase and tensin homolog (PTEN), phospholipase C (PLC), inositol 1,4,5-trisphosphate (IP3) receptor-like protein A (IplA), polyphosphate kinase 1 (Ppk1), or the TOR complex 2 component PiaA have significantly reduced sensitivity to polyphosphate-induced proliferation inhibition. Polyphosphate upregulates IP3, and this requires GrlD, GefA, PTEN, PLC, and PiaA. Polyphosphate also upregulates cytosolic Ca2+, and this requires GrlD, Gβ, GefA, RasC, PLC, IplA, Ppk1, and PiaA. Together, these data suggest that polyphosphate uses signal transduction pathways including IP3/Ca2+ to inhibit the proliferation of D. discoideum. IMPORTANCE Many mammalian tissues such as the liver have the remarkable ability to regulate their size and have their cells stop proliferating when the tissue reaches the correct size. One possible mechanism involves the cells secreting a signal that they all sense, and a high level of the signal tells the cells that there are enough of them and to stop proliferating. Although regulating such mechanisms could be useful to regulate tissue size to control cancer or birth defects, little is known about such systems. Here, we use a microbial system to study such a mechanism, and we find that key elements of the mechanism have similarities to human proteins. This then suggests the possibility that we may eventually be able to regulate the proliferation of selected cell types in humans and animals.
Collapse
|
4
|
Ouyang C, Li J, Zheng X, Mu J, Torres G, Wang Q, Zou MH, Xie Z. Deletion of Ulk1 inhibits neointima formation by enhancing KAT2A/GCN5-mediated acetylation of TUBA/α-tubulin in vivo. Autophagy 2021; 17:4305-4322. [PMID: 33985412 DOI: 10.1080/15548627.2021.1911018] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
ULK1 (unc-51 like autophagy activating kinase) has a central role in initiating macroautophagy/autophagy, a process that contributes to atherosclerosis and neointima hyperplasia, or excessive tissue growth that leads to vessel dysfunction. However, the role of ULK1 in neointima formation remains unclear. We aimed to determine how Ulk1 deletion affected neointima formation and to investigate the underlying mechanisms. We measured autophagy activity, vascular smooth muscle cell (VSMC) migration and neointima hyperplasia in cultured VSMCs and ligation-injured mouse carotid arteries from male wild-type (WT, C57BL/6 J) and VSMC-specific ulk1 knockout (ulk1 KO) mice. Carotid artery ligation in WT mice increased ULK1 protein expression, and concurrently increased autophagic flux and neointima formation. Treating human aortic smooth muscle cells (HASMCs) with PDGF (platelet derived growth factor) increased ULK1 expression, activated autophagy, and promoted cell migration. Further, smooth muscle cell-specific deletion of Ulk1 suppressed autophagy, inhibited VSMC migration, and impeded neointima hyperplasia. Mechanistically, Ulk1 deletion inhibited autophagic degradation of histone acetyltransferase protein KAT2A/GCN5 (K[lysine] acetyltransferase 2A), resulting in accumulation of KAT2A that directly acetylated TUBA/α-tubulin and subsequently increased protein levels of acetylated TUBA. The acetylation of TUBA increased microtubule stability and inhibited VSMC directional migration and neointima formation. Finally, local transfection of Kat2a siRNA decreased TUBA acetylation and prevented the attenuation of vascular injury-induced neointima formation in ulk1 KO mice. These findings suggest that Ulk1 deletion inhibits neointima formation by reducing autophagic degradation of KAT2A and increasing TUBA acetylation in VSMCs.Abbreviations: ACTA2/α-SMA: actin, alpha 2, smooth muscle, aorta; ACTB: actin beta; ATAT1: alpha tubulin acetyltransferase 1; ATG: autophagy related; BECN1: beclin 1; BP: blood pressure; CAL: carotid artery ligation; CQ: chloroquine diphosphate; EC: endothelial cells; EEL: external elastic layer; FBS: fetal bovine serum; GAPDH: glyceraldehyde 3-phosphate dehydrogenase; HASMCs: human aortic smooth muscle cells; HAT1: histone acetyltransferase 1; HDAC: histone deacetylase; IEL: inner elastic layer; IP: immunoprecipitation; KAT2A/GCN5: K(lysine) acetyltransferase 2A; KAT8/hMOF: lysine acetyltransferase 8; MAP1LC3: microtubule associated protein 1 light chain 3; MYH11: myosin heavy chain 11; PBS: phosphate-buffered saline; PDGF: platelet derived growth factor; PECAM1/CD31: platelet and endothelial cell adhesion molecule 1; RAC3: Rac family small GTPase 3; SIRT2: sirtuin 2; SPP1/OPN: secreted phosphoprotein 1; SQSTM1/p62: sequestosome 1; TAGLN/SM22: transgelin; TUBA: tubulin alpha; ULK1: unc-51 like autophagy activating kinase; VSMC: vascular smooth muscle cell; VVG: Verhoeff Van Gieson; WT: wild type.
Collapse
Affiliation(s)
- Changhan Ouyang
- Hubei Key Laboratory of Cardiovascular, Cerebrovascular and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China
| | - Jian Li
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Xiaoxu Zheng
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Jing Mu
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Gloria Torres
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Qilong Wang
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Ming-Hui Zou
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Zhonglin Xie
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| |
Collapse
|
5
|
Cutruzzolà A, Irace C, Frazzetto M, Sabatino J, Gullace R, De Rosa S, Spaccarotella C, Concolino D, Indolfi C, Gnasso A. Functional and morphological cardiovascular alterations associated with neurofibromatosis 1. Sci Rep 2020; 10:12070. [PMID: 32694667 PMCID: PMC7374589 DOI: 10.1038/s41598-020-68908-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/30/2020] [Indexed: 11/26/2022] Open
Abstract
Subjects with Neurofibromatosis 1 (NF1) develop vascular complications. The protein product of the gene affected in NF1, neurofibromin, physiologically modulates endothelial function and preserves vascular and myocardial structure. Our study aimed to verify whether subjects with NF1 have early, preclinical abnormalities of carotid artery structure, brachial artery function, and cardiac function. We recruited 22 NF1 subjects without previous cardiovascular events and 22 healthy control subjects. All subjects underwent measurement of carotid artery intima-media thickness (IMT), evaluation of brachial artery endothelial function after ischemia and exercise, and cardiac function. Mean IMT was 543 ± 115 μ in NF1 subjects and 487 ± 70 μ in Controls (p < 0.01). Endothelial function was significantly dumped in NF1 subjects. The dilation after ischemia and exercise was respectively 7.5(± 4.8)% and 6.7(± 3.0)% in NF1 versus 10.5(± 1.2)% and 10.5(± 2.1)% in control subjects (p < 0.02; p < 0.002). Left ventricular systolic function assessed by Global Longitudinal Strain was significantly different between NF1 subjects and Controls: − 19.3(± 1.7)% versus − 21.5(± 2.7)% (p < 0.008). These findings demonstrate that NF1 patients have early morphological and functional abnormalities of peripheral arteries and systolic cardiac impairment and suggest the need for a tight cardiovascular risk evaluation and primary prevention in subjects with NF1.
Collapse
Affiliation(s)
- Antonio Cutruzzolà
- Dipartimento di Medicina Sperimentale e Clinica, University Magna Græcia, Viale Europa Località Germaneto, 88100, Catanzaro, Italy
| | - Concetta Irace
- Dipartimento di Scienze della Salute, University Magna Græcia, Catanzaro, Italy
| | - Marco Frazzetto
- Dipartimento di Scienze della Salute, University Magna Græcia, Catanzaro, Italy
| | - Jolanda Sabatino
- Dipartimento di Scienze Mediche e Chirurgiche, University Magna Græcia, Catanzaro, Italy.,Center of Cardiovascular Research, University Magna Graecia, Mediterranea Cardio Centro, Catanzaro, Napoli, Italy
| | - Rosa Gullace
- Dipartimento di Scienze della Salute, University Magna Græcia, Catanzaro, Italy
| | - Salvatore De Rosa
- Dipartimento di Scienze Mediche e Chirurgiche, University Magna Græcia, Catanzaro, Italy.,Center of Cardiovascular Research, University Magna Graecia, Mediterranea Cardio Centro, Catanzaro, Napoli, Italy
| | - Carmen Spaccarotella
- Center of Cardiovascular Research, University Magna Graecia, Mediterranea Cardio Centro, Catanzaro, Napoli, Italy
| | - Daniela Concolino
- Dipartimento di Scienze della Salute, University Magna Græcia, Catanzaro, Italy
| | - Ciro Indolfi
- Dipartimento di Scienze Mediche e Chirurgiche, University Magna Græcia, Catanzaro, Italy.,Center of Cardiovascular Research, University Magna Graecia, Mediterranea Cardio Centro, Catanzaro, Napoli, Italy
| | - Agostino Gnasso
- Dipartimento di Medicina Sperimentale e Clinica, University Magna Græcia, Viale Europa Località Germaneto, 88100, Catanzaro, Italy.
| |
Collapse
|
6
|
Zhu Y, Zhang H, Zhang Y, Wu H, Wei L, Zhou G, Zhang Y, Deng L, Cheng Y, Li M, Santos HA, Cui W. Endovascular Metal Devices for the Treatment of Cerebrovascular Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1805452. [PMID: 30589125 DOI: 10.1002/adma.201805452] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/20/2018] [Indexed: 06/09/2023]
Abstract
Cerebrovascular disease involves various medical disorders that obstruct brain blood vessels or deteriorate cerebral circulation, resulting in ischemic or hemorrhagic stroke. Nowadays, platinum coils with or without biological modification have become routine embolization devices to reduce the risk of cerebral aneurysm bleeding. Additionally, many intracranial stents, flow diverters, and stent retrievers have been invented with uniquely designed structures. To accelerate the translation of these devices into clinical usage, an in-depth understanding of the mechanical and material performance of these metal-based devices is critical. However, considering the more distal location and tortuous anatomic characteristics of cerebral arteries, present devices still risk failing to arrive at target lesions. Consequently, more flexible endovascular devices and novel designs are under urgent demand to overcome the deficiencies of existing devices. Herein, the pros and cons of the current structural designs are discussed when these devices are applied to the treatment of diseases ranging broadly from hemorrhages to ischemic strokes, in order to encourage further development of such kind of devices and investigation of their use in the clinic. Moreover, novel biodegradable materials and drug elution techniques, and the design, safety, and efficacy of personalized devices for further clinical applications in cerebral vasculature are discussed.
Collapse
Affiliation(s)
- Yueqi Zhu
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Shanghai, 200233, P. R. China
| | - Hongbo Zhang
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
- Department of Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, FI-20520, Finland
- Turku Center for Biotechnology, University of Turku and Åbo Akademi University, Turku, FI-20520, Finland
| | - Yiran Zhang
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Shanghai, 200233, P. R. China
| | - Huayin Wu
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Liming Wei
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Shanghai, 200233, P. R. China
| | - Gen Zhou
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Shanghai, 200233, P. R. China
| | - Yuezhou Zhang
- Department of Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, FI-20520, Finland
- Turku Center for Biotechnology, University of Turku and Åbo Akademi University, Turku, FI-20520, Finland
| | - Lianfu Deng
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Yingsheng Cheng
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Shanghai, 200233, P. R. China
| | - Minghua Li
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan Road, Shanghai, 200233, P. R. China
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
- Helsinki Institute of Life Science, University of Helsinki, FI-00014, Helsinki, Finland
| | - Wenguo Cui
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| |
Collapse
|
7
|
Hall S, Agrawal DK. Delivery of viral vectors for gene therapy in intimal hyperplasia and restenosis in atherosclerotic swine. Drug Deliv Transl Res 2018; 8:918-927. [PMID: 28707263 DOI: 10.1007/s13346-017-0409-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiovascular diseases including atherosclerosis are a major financial and health burden globally. Inflammation associated with atherosclerosis results in the development of plaques that can rupture causing thrombosis, stroke, or death. The most widely used treatment for the removal of atherosclerotic plaques is percutaneous transluminal coronary angioplasty (PTCA) with or without stenting. Although this is a safer and minimally invasive method, restenosis and intimal hyperplasia after interventional procedure remains a major hurdle and more refined approaches are needed. Studies in large animal models such as pigs have facilitated a greater understanding of the underlying mechanisms of the disease and provided novel targets for therapeutic intervention. In pre-clinical studies, viral vector gene therapy has emerged as a promising option for the reduction and/or prevention of restenosis and intimal hyperplasia. Although studies in animal models have generated promising results, clinical trials have yet to prove the clinical efficacy of gene therapy in coronary artery diseases. In this review, we examined and critically reviewed the most recent advances in viral vector gene therapy obtained from studies using porcine model of atherosclerosis.
Collapse
Affiliation(s)
- Sannette Hall
- Department of Clinical and Translational Science, School of Medicine, Creighton University, Omaha, NE, 68178, USA
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, School of Medicine, Creighton University, Omaha, NE, 68178, USA. .,Department of Clinical and Translational Science, The Peekie Nash Carpenter Endowed Chair in Medicine, School of Medicine, Creighton University, CRISS II Room 510, 2500 California Plaza, Omaha, NE, 68178, USA.
| |
Collapse
|
8
|
RAS mutations in acute myeloid leukaemia patients: A review and meta-analysis. Clin Chim Acta 2018; 489:254-260. [PMID: 30194935 DOI: 10.1016/j.cca.2018.08.040] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 08/23/2018] [Accepted: 08/24/2018] [Indexed: 12/20/2022]
Abstract
RAS oncogene mutations frequently occur in acute myeloid leukaemia (AML), but the prognostic significance of RAS mutations in AML is inconclusive. We searched the databases of PubMed, Web of Science, EMBASE, and Cochrane from 1990 to 2018. In this study, 24 eligible studies were included, and the meta-analysis was conducted with the Comprehensive Meta-Analysis Version 2 software program. The row hazard ratio (HR) was adjusted and re-evaluated when publication bias existed after detecting all the heterogeneities. A combined analysis showed that RAS mutations were not associated with a poor prognosis in general AML patients (HR: 0.96, 95% CI: 0.78-1.19, p = 0.70). To further verify the results, a subgroup analysis was conducted. Interestingly, in the analysis of age bracket, children with RAS mutations had an unfavourable survival (HR: 1.35, 95% CI: 1.05-1.75, p = 0.02) of AML, but the adults did not (HR: 0.87, 95% CI: 0.70-1.09, p = 0.21). Further analysis of the subgroup of children indicated that patients with NRAS mutations had an adverse prognosis (HR: 1.55, 95% CI: 1.13-2.12, p = 0.007), but not those with KRAS mutations (HR: 1.51, 95% CI: 0.34-6.73, p = 0.59). In conclusion, this study revealed that RAS mutations did not influence the over survival for adults with AML. However, NRAS mutations may be a key prognostic marker related with poor survival for children with AML.
Collapse
|
9
|
Yang J, Zeng P, Yang J, Liu X, Ding J, Wang H, Chen L. MicroRNA-24 regulates vascular remodeling via inhibiting PDGF-BB pathway in diabetic rat model. Gene 2018; 659:67-76. [PMID: 29559348 DOI: 10.1016/j.gene.2018.03.056] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/11/2018] [Accepted: 03/16/2018] [Indexed: 12/26/2022]
Abstract
PURPOSE Hyperglycemia is the high risk factor of vascular remodeling induced by angioplasty, and neointimal hyperplasia is strongly implicated in the pathogenesis of vascular remodeling caused by carotid artery balloon injury. Studies have shown that MicroRNA 24 (miR-24) plays an important role in angiocardiopathy, However, the role of miR-24 is far from thorough research. In this study, we investigate whether up-regulation of miR-24 by using miR-24 recombinant adenovirus (Ad-miR-24-GFP) can inhibit PDGF-BB signaling pathway and attenuate vascular remodeling in the diabetic rat model. METHODS Male Sprague-Dawley rats (n = 60) were randomly divided into 5 groups and fed with high sugar and high fat diet (Sham, Saline, Scramble, Ad-miR-24 groups), or ordinary diet (Control group). The front four groups were treated with streptozotocin (STZ) four weeks later and the blood glucose level was closely monitored. After the successful establishment of diabetic rats, the external carotid artery was injured by pressuring balloon 1.5 after internal carotid artery ligation, then the blood vessels were harvested 14 days later and indexes were detected including the following: HE staining for the level of vascular intima thickness, immunohistochemical detection for PCNA and P27 to test the proliferative degree of vascular smooth muscle cells (VSMCs), qRT-PCR for the level of miR-24, RAS,PDGF-R, western blot for the protein levels of JNK1/2, p- JNK1/2, ERK1/2, p-ERK1/2, RAS, PDGF-R, AP-1,P27 and PCNA. Serological detection was conducted for TNF-α, IL-6, IL-8. RESULTS The delivery of Ad-miR-24 into balloon injury site has significantly increased the level of miR-24. Up-regulation of miR-24 could regulate vascular remodeling effectively, lower the level of inflammatory factors, inhibit the expression of mRNA and protein levels of JNK1/2, ERK1/2, RAS, PDGF-R, AP-1, P27, PCNA. CONCLUSION miR-24 can inhibit the expression of AP-1 via the inhibition of PDGF-BB signaling pathway, thus inhibit VSMCs proliferation and vascular remodeling.
Collapse
Affiliation(s)
- Jian Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China; Key Laboratory of Ischemic Cardiovascular and Cerebrovascular Disease Translational Medicine, Three Gorges University, China.
| | - Ping Zeng
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, China
| | - Jun Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China; Key Laboratory of Ischemic Cardiovascular and Cerebrovascular Disease Translational Medicine, Three Gorges University, China
| | - Xiaowen Liu
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China; Key Laboratory of Ischemic Cardiovascular and Cerebrovascular Disease Translational Medicine, Three Gorges University, China
| | - Jiawang Ding
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China; Key Laboratory of Ischemic Cardiovascular and Cerebrovascular Disease Translational Medicine, Three Gorges University, China
| | - Huibo Wang
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, China
| | - Lihua Chen
- Department of Optometry and Ophthalmology, Yichang Central People's Hospital, Yichang, China
| |
Collapse
|
10
|
Yu MH, Lin MC, Huang CN, Chan KC, Wang CJ. Acarbose inhibits the proliferation and migration of vascular smooth muscle cells via targeting Ras signaling. Vascul Pharmacol 2018; 103-105:8-15. [PMID: 29432898 DOI: 10.1016/j.vph.2018.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/22/2017] [Accepted: 02/06/2018] [Indexed: 12/31/2022]
Abstract
Atherosclerosis involves the proliferation and migration of vascular smooth muscle cells (VSMCs). The migration of VSMCs from the media into the intima and their subsequent proliferation are important processes in neointima formation in atherosclerosis and restenosis after percutaneous coronary interventions. Acarbose, an alpha-glucosidase inhibitor, has been demonstrated to not affect serum levels of glucose and decrease the progression of intima-media thickening in rabbits fed with a high cholesterol diet (HCD). We previously showed that increased Ras protein levels enhanced the migration of TNF-α treated A7r5 cells. The aim of this study was to determine the inhibitory effects of acarbose on Ras expression in A7r5 cells. Acarbose also inhibited the phosphorylation of focal adhesion kinase (FAK) and Akt, activities of the matrix metalloproteinases (MMPs) MMP-2 and MMP-9, and protein expressions of small G proteins (Ras, Cdc42, RhoA, and Rac1) in a dose-dependent manner. We also found that acarbose could effectively inhibit the proliferation and migration of RasG12V A7r5 cells by blocking small G proteins and phosphoinositide-3-kinase (PI3K)/Akt signaling. These studies demonstrated that acarbose could theoretically decrease atherosclerosis by targeting Ras signaling.
Collapse
Affiliation(s)
- Meng-Hsun Yu
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung 402, Taiwan
| | - Ming-Cheng Lin
- Department of Internal Medicine, Chung-Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Road, Taichung 402, Taiwan; School of Medicine, Institute of Medicine, Chung-Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung 402, Taiwan
| | - Chien-Ning Huang
- Department of Internal Medicine, Chung-Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Road, Taichung 402, Taiwan; School of Medicine, Institute of Medicine, Chung-Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung 402, Taiwan
| | - Kuei-Chuan Chan
- Department of Internal Medicine, Chung-Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Road, Taichung 402, Taiwan; School of Medicine, Institute of Medicine, Chung-Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung 402, Taiwan.
| | - Chau-Jong Wang
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung 402, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Road, Taichung 402, Taiwan.
| |
Collapse
|
11
|
Sorrentino S, Iaconetti C, De Rosa S, Polimeni A, Sabatino J, Gareri C, Passafaro F, Mancuso T, Tammè L, Mignogna C, Camastra C, Esposito G, Curcio A, Torella D, Indolfi C. Hindlimb Ischemia Impairs Endothelial Recovery and Increases Neointimal Proliferation in the Carotid Artery. Sci Rep 2018; 8:761. [PMID: 29335599 PMCID: PMC5768880 DOI: 10.1038/s41598-017-19136-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 12/18/2017] [Indexed: 01/29/2023] Open
Abstract
Peripheral ischemia is associated with higher degree of endothelial dysfunction and a worse prognosis after percutaneous coronary interventions (PCI). However, the role of peripheral ischemia on vascular remodeling in remote districts remains poorly understood. Here we show that the presence of hindlimb ischemia significantly enhances neointima formation and impairs endothelial recovery in balloon-injured carotid arteries. Endothelial-derived microRNAs are involved in the modulation of these processes. Indeed, endothelial miR-16 is remarkably upregulated after vascular injury in the presences of hindlimb ischemia and exerts a negative effect on endothelial repair through the inhibition of RhoGDIα and nitric oxide (NO) production. We showed that the repression of RhoGDIα by means of miR-16 induces RhoA, with consequent reduction of NO bioavailability. Thus, hindlimb ischemia affects negative carotid remodeling increasing neointima formation after injury, while systemic antagonizzation of miR-16 is able to prevent these negative effects.
Collapse
Affiliation(s)
- Sabato Sorrentino
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Claudio Iaconetti
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Salvatore De Rosa
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Alberto Polimeni
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Jolanda Sabatino
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Clarice Gareri
- Department of Medicine, Duke University, Durham, 27710, NC, USA
| | - Francesco Passafaro
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Teresa Mancuso
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Laura Tammè
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Chiara Mignogna
- Department of Health Science, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Caterina Camastra
- Department of Health Science, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Giovanni Esposito
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Antonio Curcio
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Daniele Torella
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Ciro Indolfi
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy. .,URT-CNR of IFC, Magna Graecia University, Catanzaro, Italy.
| |
Collapse
|
12
|
Stopeck AT, Vahedian M, Williams SK. Transfer and Expression of the Interferon Gamma Gene in Human Endothelial Cells Inhibits Vascular Smooth Muscle Cell Growth in Vitro. Cell Transplant 2017; 6:1-8. [PMID: 9040949 DOI: 10.1177/096368979700600103] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Intimal hyperplasia in blood vessels is primarily caused by the migration and proliferation of vascular smooth muscle cells. Excessive intimal thickening characterizes atherosclerosis as well as bypass graft and angioplasty failures. Endothelial cell-smooth muscle cell interactions and local cytokine production are important regulators of smooth muscle cell growth. Interferon gamma (γ-IFN), a product of T lymphocytes found in atherosclerotic lesions, inhibits smooth muscle cell proliferation in vitro. To determine if local delivery of γ-IFN may be useful in the treatment or prevention of vascular proliferative diseases, we transferred the human γ-IFN gene into endothelial cells isolated from human arteries and microvessels using a retroviral vector. Biologically active γ-IFN was produced and secreted by γ-IFN transduced endothelial cells, but not by control, nontransduced cells, or cells identically transduced with E. coli beta galactosidase (β-gal). To more closely approximate the microenvironment of blood vessels, subconfluent smooth muscle cells were plated in coculture with control, nontransduced endothelial cells, γ-IFN transduced endothelial cells, or β-gal transduced endothelial cells. Smooth muscle cell growth was inhibited 30-70% by coculture with γ-IFN transduced endothelial cells compared to coculture with β-gal transduced or control endothelial cells (p < 0.05). Our results suggest endothelial cells modified to produce γ-IFN may be a useful therapy in proliferative vascular diseases. Copyright © 1997 Elsevier Science Inc.
Collapse
Affiliation(s)
- A T Stopeck
- Section of Hematology/Oncology, Arizona Cancer Center, University of Arizona College of Medicine, Tucson 85724, USA
| | | | | |
Collapse
|
13
|
Chandiwal A, Balasubramanian V, Baldwin ZK, Conte MS, Schwartz LB. Gene Therapy for the Extension of Vein Graft Patency: A Review. Vasc Endovascular Surg 2016; 39:1-14. [PMID: 15696243 DOI: 10.1177/153857440503900101] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The mainstay of treatment for long-segment small-vessel chronic occlusive disease not amenable to endovascular intervention remains surgical bypass grafting using autologous vein. The procedure is largely successful and the immediate operative results almost always favorable. However, the lifespan of a given vein graft is highly variable, and less than 50% will remain primarily patent after 5 years. The slow process of graft malfunction is a result of the vein's chronic maladaptive response to the systemic arterial environment, its primary component being the uncontrolled proliferation of vascular smooth muscle cells (SMCs). It has recently been suggested that this response might be attenuated through pre-implantation genetic modification of the vein, so-called gene therapy for the extension of vein graft patency. Gene therapy seems particularly well suited for the prevention or postponement of vein graft failure since: (1) the stimulation of SMC proliferation appears to largely be an early and transient process, matching the kinetics of current gene transfer technology; (2) most veins are relatively normal and free of disease at the time of bypass allowing for effective gene transfer using a variety of systems; and (3) the target tissue is directly accessible during operation because manipulation and irrigation of the vein is part of the normal workflow of the surgical procedure. This review briefly summarizes the current knowledge of the incidence and basic mechanisms of vein graft failure, the vector systems and molecular targets that have been proposed as possible pre-treatments, the results of experimental genetic modification of vein grafts, and the few available clinical studies of gene therapy for vascular proliferative disorders.
Collapse
Affiliation(s)
- Amito Chandiwal
- Section of Vascular Surgery, Department of Surgery, University of Chicago, IL 60637, USA
| | | | | | | | | |
Collapse
|
14
|
Iaconetti C, De Rosa S, Polimeni A, Sorrentino S, Gareri C, Carino A, Sabatino J, Colangelo M, Curcio A, Indolfi C. Down-regulation of miR-23b induces phenotypic switching of vascular smooth muscle cellsin vitroandin vivo. Cardiovasc Res 2015; 107:522-33. [DOI: 10.1093/cvr/cvv141] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 04/22/2015] [Indexed: 12/22/2022] Open
|
15
|
Wang LF, Tao LW, Huang MX, Liao WB, Zhu YZ, Zhou WB, Li H, Li D, Lu HT, Zhang BZ, Chen Z. Clinical Evaluation of Coronary In-Stent Restenosis Using Dual-Source Computed Tomography. Echocardiography 2015; 32:1681-7. [PMID: 25816917 DOI: 10.1111/echo.12932] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Lian-Fa Wang
- Department of Cardiology; People's Liberation Army No. 105 Hospital; Hefei Anhui China
| | - Li-Wei Tao
- Department of Cardiothoracic Surgery; The Second People's Hospital of Fuyang City; Fuyang Anhui China
| | - Meng-Xun Huang
- Department of Cardiology; People's Liberation Army No. 105 Hospital; Hefei Anhui China
| | - Wen-Bin Liao
- Department of Cardiology; People's Liberation Army No. 105 Hospital; Hefei Anhui China
| | - You-Zhi Zhu
- Department of Radiology; People's Liberation Army No. 105 Hospital; Hefei Anhui China
| | - Wen-Bing Zhou
- Department of Cardiology; People's Liberation Army No. 105 Hospital; Hefei Anhui China
| | - Hua Li
- Department of Cardiology; People's Liberation Army No. 105 Hospital; Hefei Anhui China
| | - Dan Li
- Department of Cardiology; People's Liberation Army No. 105 Hospital; Hefei Anhui China
| | - Hong-Tao Lu
- Department of Cardiology; People's Liberation Army No. 105 Hospital; Hefei Anhui China
| | - Bang-Zhu Zhang
- Department of Cardiology; People's Liberation Army No. 105 Hospital; Hefei Anhui China
| | - Zhen Chen
- Department of Cardiology; People's Liberation Army No. 105 Hospital; Hefei Anhui China
| |
Collapse
|
16
|
De Rosa S, Indolfi C. Circulating microRNAs as Biomarkers in Cardiovascular Diseases. EXPERIENTIA SUPPLEMENTUM (2012) 2015; 106:139-149. [PMID: 26608202 DOI: 10.1007/978-3-0348-0955-9_6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
MicroRNAs, key regulators of biological processes, are involved in the pathophysiological mechanisms underlying human diseases, including cardiovascular diseases. Their recent discovery revealed a previously unknown layer of pathophysiologic regulators, which also play a key role in the regulation of several aspects of cardiovascular diseases. More recently, it was demonstrated that circulating microRNAs can be measured in the blood. Hence, the potential use of microRNAs as disease biomarkers attracted many research groups. Indeed, their unusual stability in the bloodstream and during prolonged storage make circulating miRs very interesting as potential biomarkers. Circulating microRNAs are emerging as the next generation "smart" biomarkers and could be helpful in further improving the diagnostic and therapeutic processes of cardiovascular diseases. The present chapter summarizes the most relevant experimental evidence on circulating microRNAs in cardiovascular diseases, including arterial remodeling, restenosis, coronary artery disease, acute coronary syndromes, hypertension, heart failure, and ischemic stroke, highlighting potential pathophysiological correlations to the mechanisms underlying cardiovascular diseases.
Collapse
Affiliation(s)
- Salvatore De Rosa
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Ciro Indolfi
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy. .,URT-CNR, Magna Graecia University, Catanzaro, 88100, Italy.
| |
Collapse
|
17
|
Yiqihuoxuejiedu formula inhibits vascular remodeling by reducing proliferation and secretion of adventitial fibroblast after balloon injury. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:849167. [PMID: 24987435 PMCID: PMC4058465 DOI: 10.1155/2014/849167] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 03/28/2014] [Accepted: 04/30/2014] [Indexed: 11/18/2022]
Abstract
Vascular remodeling occurs in atherosclerosis, hypertension, and restenosis after percutaneous coronary intervention. Adventitial remodeling may be a potential therapeutic target. Yiqihuoxuejiedu formula uses therapeutic principles from Chinese medicine to supplement Qi, activate blood circulation, and resolve toxin and it has been shown to inhibit vascular stenosis. To investigate effects and mechanisms of the formula on inhibiting vascular remodeling, especially adventitial remodeling, rats with a balloon injury to their common carotid artery were used and were treated for 7 or 28 days after injury. The adventitial area and α -SMA expression increased at 7 days after injury, which indicated activation and proliferation of adventitial fibroblasts. Yiqihuoxuejiedu formula reduced the adventitial areas at 7 days, attenuated the neointima and vessel wall area, stenosis percent, and α -SMA expression in the neointima, and reduced collagen content and type I/III collagen ratio in the adventitia at 28 days. Yiqihuoxuejiedu formula had more positive effects than Captopril in reducing intimal proliferation and diminishing stenosis, although Captopril lowered neointimal α -SMA expression and reduced the collagen content at 28 days. Yiqihuoxuejiedu formula has inhibitory effects on positive and negative remodeling by reducing adventitial and neointimal proliferation, reducing content, and elevating adventitial compliance.
Collapse
|
18
|
Bhardwaj S, Roy H, Ylä-Herttuala S. Gene therapy to prevent occlusion of venous bypass grafts. Expert Rev Cardiovasc Ther 2014; 6:641-52. [DOI: 10.1586/14779072.6.5.641] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
19
|
Affiliation(s)
- Salvatore De Rosa
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University
| | - Antonio Curcio
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University
| | - Ciro Indolfi
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University
- URT-CNR, Magna Graecia University
| |
Collapse
|
20
|
Non-coding RNAs: the "dark matter" of cardiovascular pathophysiology. Int J Mol Sci 2013; 14:19987-20018. [PMID: 24113581 PMCID: PMC3821599 DOI: 10.3390/ijms141019987] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 09/12/2013] [Accepted: 09/16/2013] [Indexed: 12/17/2022] Open
Abstract
Large-scale analyses of mammalian transcriptomes have identified a significant number of different RNA molecules that are not translated into protein. In fact, the use of new sequencing technologies has identified that most of the genome is transcribed, producing a heterogeneous population of RNAs which do not encode for proteins (ncRNAs). Emerging data suggest that these transcripts influence the development of cardiovascular disease. The best characterized non-coding RNA family is represented by short highly conserved RNA molecules, termed microRNAs (miRNAs), which mediate a process of mRNA silencing through transcript degradation or translational repression. These microRNAs (miRNAs) are expressed in cardiovascular tissues and play key roles in many cardiovascular pathologies, such as coronary artery disease (CAD) and heart failure (HF). Potential links between other ncRNAs, like long non-coding RNA, and cardiovascular disease are intriguing but the functions of these transcripts are largely unknown. Thus, the functional characterization of ncRNAs is essential to improve the overall understanding of cellular processes involved in cardiovascular diseases in order to define new therapeutic strategies. This review outlines the current knowledge of the different ncRNA classes and summarizes their role in cardiovascular development and disease.
Collapse
|
21
|
Loirand G, Sauzeau V, Pacaud P. Small G Proteins in the Cardiovascular System: Physiological and Pathological Aspects. Physiol Rev 2013; 93:1659-720. [DOI: 10.1152/physrev.00021.2012] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Small G proteins exist in eukaryotes from yeast to human and constitute the Ras superfamily comprising more than 100 members. This superfamily is structurally classified into five families: the Ras, Rho, Rab, Arf, and Ran families that control a wide variety of cell and biological functions through highly coordinated regulation processes. Increasing evidence has accumulated to identify small G proteins and their regulators as key players of the cardiovascular physiology that control a large panel of cardiac (heart rhythm, contraction, hypertrophy) and vascular functions (angiogenesis, vascular permeability, vasoconstriction). Indeed, basal Ras protein activity is required for homeostatic functions in physiological conditions, but sustained overactivation of Ras proteins or spatiotemporal dysregulation of Ras signaling pathways has pathological consequences in the cardiovascular system. The primary object of this review is to provide a comprehensive overview of the current progress in our understanding of the role of small G proteins and their regulators in cardiovascular physiology and pathologies.
Collapse
Affiliation(s)
- Gervaise Loirand
- INSERM, UMR S1087; University of Nantes; and CHU Nantes, l'Institut du Thorax, Nantes, France
| | - Vincent Sauzeau
- INSERM, UMR S1087; University of Nantes; and CHU Nantes, l'Institut du Thorax, Nantes, France
| | - Pierre Pacaud
- INSERM, UMR S1087; University of Nantes; and CHU Nantes, l'Institut du Thorax, Nantes, France
| |
Collapse
|
22
|
Rodriguez MP, Emond ZM, Varu VN, Ahanchi SS, Martinez J, Kibbe MR. Nitric oxide differentially affects ERK and Akt in type 1 and type 2 diabetic rats. J Surg Res 2013; 183:944-951. [PMID: 23608617 DOI: 10.1016/j.jss.2013.02.055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 02/20/2013] [Accepted: 02/27/2013] [Indexed: 11/26/2022]
Abstract
BACKGROUND We have shown that nitric oxide (NO) is more effective at inhibiting neointimal hyperplasia in type 2 diabetic rats than in nondiabetic rats, but is not effective in type 1 diabetic rats. Insulin signaling is mediated by the ERK and Akt pathways, and thus we hypothesized that NO differentially affects ERK and Akt activity in type 1 versus type 2 diabetic rats. MATERIALS AND METHODS To investigate this hypothesis, we induced type 2 diabetes in Zucker diabetic fatty (ZDF) rats by feeding them Purina 5008 chow. To induce type 1 diabetes, lean Zucker (LZ) rats were injected with streptozotocin (STZ; 60 mg/kg). The carotid artery injury model was performed. Groups included injury and injury + PROLI/NO (20 mg/kg) (n = 6/group). RESULTS Three days following injury, all animal models exhibited an increase in pERK levels. Whereas NO reduced pERK levels in LZ and STZ rats, NO had no effect on pERK levels in ZDF rats. Following a similar pattern, NO reduced pAkt levels in LZ and STZ rats but increased pAkt levels in ZDF rats. Fourteen days following injury, NO increased total pERK levels throughout the arterial wall in both the STZ and ZDF rats. These changes were greatest in the adventitia. Interestingly, whereas NO decreased total pAkt levels in LZ and STZ rats, NO increased pAkt levels in ZDF rats. Evaluation of the pERK:pAkt ratio revealed that NO increased this ratio in LZ and STZ rats but decreased the ratio in ZDF rats. CONCLUSIONS We report that NO differentially affects the expression of pERK and pAkt in type 1 versus type 2 diabetic rats. Given that NO is more effective at inhibiting neointimal hyperplasia in type 2 diabetic animals, the pERK:pAkt ratio may be the best surrogate to predict efficacy.
Collapse
Affiliation(s)
- Monica P Rodriguez
- Division of Vascular Surgery and Institute for BioNanotechnology in Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Zachary M Emond
- Division of Vascular Surgery and Institute for BioNanotechnology in Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Vinit N Varu
- Division of Vascular Surgery and Institute for BioNanotechnology in Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Sadaf S Ahanchi
- Division of Vascular Surgery and Institute for BioNanotechnology in Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Janet Martinez
- Division of Vascular Surgery and Institute for BioNanotechnology in Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Jesse Brown VA Medical Center, Chicago, Illinois
| | - Melina R Kibbe
- Division of Vascular Surgery and Institute for BioNanotechnology in Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Jesse Brown VA Medical Center, Chicago, Illinois
| |
Collapse
|
23
|
Vascular miRNAs After Balloon Angioplasty. Trends Cardiovasc Med 2013; 23:9-14. [DOI: 10.1016/j.tcm.2012.08.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 08/09/2012] [Accepted: 08/09/2012] [Indexed: 11/21/2022]
|
24
|
Abstract
Background Lower extremity artery disease (LE-PAD) is one of the most common manifestations of atherosclerosis, particularly in elderly patients, and it is related to a high cardiovascular risk. Description It is well established that statin therapy is characterized by crucial benefits on cardiovascular system by limiting atherosclerotic progression and reducing cardiovascular events and mortality. A growing body of evidence support efficacy of statins in LE-PAD due to the ability of both reducing cardiovascular risk and improving walking distance and, hence, quality of life. Consequently, statin therapy should be considered in all LE-PAD patients and new LDL-cholesterol targets should be reached. Conclusions Our opinion is that statin therapy remains still underutilized or with inadequate dosage, so therapy of LE-PAD patients should be improved to obtain all the demonstrated benefits of statins.
Collapse
|
25
|
Franzone A, Ferrone M, Carotenuto G, Carbone A, Scudiero L, Serino F, Scudiero F, Izzo R, Piccolo R, Saviano S, Amato B, Perrino C, Trimarco B, Esposito G. The role of atherectomy in the treatment of lower extremity peripheral artery disease. BMC Surg 2012; 12 Suppl 1:S13. [PMID: 23173800 PMCID: PMC3499201 DOI: 10.1186/1471-2482-12-s1-s13] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background The incidence of lower extremity peripheral artery disease (LE-PAD) continues to increase and associated morbidity remains high. Despite the significant development of percutaneous revascularization strategies, over the past decade, LE-PAD still represents a unique challenge for interventional cardiologists and vascular surgeons. Method Typical features of atherosclerosis that affects peripheral vascular bed (diffuse nature, poor distal runoff, critical limb ischemia, chronic total occlusion) contribute to the disappointing results of traditional percutaneous transluminal angioplasty (PTA). New technologies have been developed in attempt to improve the safety and effectiveness of percutaneous revascularization. Among these, atherectomy, debulking and removing atherosclerotic plaque, offers the potential advantage of eliminating stretch on arterial walls and reducing rates of restenosis. Conclusions This review summarizes the features and the current applications of new debulking devices.
Collapse
Affiliation(s)
- Anna Franzone
- Department of Clinical Medicine, Cardiovascular and Immunological Sciences, Federico II University, Via S, Pansini 5, 80131 Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Inhibition of miR-92a increases endothelial proliferation and migration in vitro as well as reduces neointimal proliferation in vivo after vascular injury. Basic Res Cardiol 2012; 107:296. [PMID: 22890560 DOI: 10.1007/s00395-012-0296-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Revised: 07/16/2012] [Accepted: 08/03/2012] [Indexed: 01/07/2023]
Abstract
The role of miR-92a on vascular remodelling after injury is currently unknown. Thus, the aim of the present study was to evaluate the role of miR-92a on rat endothelial and vascular smooth muscle cells proliferation and migration in vitro as well as after balloon injury or arterial stenting in vivo. MiR-92a was highly expressed in RAO-ECs and vascular endothelium, but not in RAO-SMCs or medial smooth muscle as assessed by real-time RT-PCR. Importantly, BrdU incorporation and wound healing assay provide evidence that functional inhibition of miR-92a resulted in an increased RAO-ECs proliferation and migration, but had no effect on RAO-SMCs proliferation or migration in vitro. Immunoblotting analysis revealed an increased phosphorylation of ERK1/2, JNK/SAPK as well as eNOS and phospho-eNOS increased expression level in RAO-ECs as a consequence of miR-92a inhibition. Using gain and loss of function experiments, we showed that miR-92a modulates regulation of KLF4 and MKK4 expression level in endothelial cells. Finally, in vivo administration of antagomiR-92a significantly enhanced re-endothelialization in injured carotid arteries and reduced neointimal formation after balloon injury or arterial stenting. These data provide the first evidence that inhibition of miR-92a may represent a novel strategy to improve endothelial regeneration and reduce restenosis after vascular injury.
Collapse
|
27
|
Bolchi C, Pallavicini M, Bernini SK, Chiodini G, Corsini A, Ferri N, Fumagalli L, Straniero V, Valoti E. Thiazole- and imidazole-containing peptidomimetic inhibitors of protein farnesyltransferase. Bioorg Med Chem Lett 2011; 21:5408-12. [DOI: 10.1016/j.bmcl.2011.07.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 06/30/2011] [Accepted: 07/03/2011] [Indexed: 11/15/2022]
|
28
|
Emond ZM, Kibbe MR. Clinical science review article: understanding the implications of diabetes on the vascular system. Vasc Endovascular Surg 2011; 45:481-9. [PMID: 21571777 DOI: 10.1177/1538574411408354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Patients with diabetes comprise an extremely complex subset of patients for the vascular surgeon. Often, they have numerous comorbidities that can further complicate matters. The diabetic environment is highly complex and the interplay of various diseases makes this an extremely challenging condition to manage. Knowing the mechanisms by which diabetes inflicts adverse microscopic changes in the vasculature allows the clinician to anticipate problems and minimize the heightened risks observed in diabetic patients undergoing surgery. In this review, we will illustrate how diabetes affects the vasculature and how the molecular and cellular derangements that occur in diabetic environments lead to these pathophysiologic consequences.
Collapse
Affiliation(s)
- Zachary M Emond
- Department of Surgery, University of Illinois at Chicago, IL, USA
| | | |
Collapse
|
29
|
Curcio A, Torella D, Indolfi C. Mechanisms of smooth muscle cell proliferation and endothelial regeneration after vascular injury and stenting: approach to therapy. Circ J 2011; 75:1287-96. [PMID: 21532177 DOI: 10.1253/circj.cj-11-0366] [Citation(s) in RCA: 189] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bare metal stents (BMS) successfully prevented abrupt artery closure and reduced the restenosis rate compared with balloon angioplasty. This review summarizes laboratory and recent clinical investigations concerning neointimal formation and endothelial regeneration after vascular injury. BMS efficacy was severely hampered by proliferating vascular smooth muscle cells (VSMCs), and the resultant neointimal hyperplasia, which is the only mechanism responsible for restenosis after metal stent placement. The advent of drug-eluting stents (DES) in 2002 have since then revolutionized interventional cardiology. By using the stent struts as a platform coated with polymers to elute drugs targeting VSMC proliferation, a substantial attenuation of in-stent restenosis is feasible. As with any medical innovation this technology still has restrictive factors, and novel approaches are promoted to improve the safety and efficacy of DES. Indeed, the antiproliferative properties of DES impair and/or delay endothelialization, hence leading to late stent thrombosis. Improvements in percutaneous coronary intervention procedures include the use of the so-called "second-generation DES", together with new coating technologies, bioabsorbable stents, and non-drug-based stent coatings. Particular emphasis will be placed on the concept that endothelial regeneration might be pursued as well as reduction of VSMC proliferation to allow stable successful revascularization after DES deployment.
Collapse
Affiliation(s)
- Antonio Curcio
- Division of Cardiology, Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy
| | | | | |
Collapse
|
30
|
Stein W, Schrepfer S, Itoh S, Kimura N, Velotta J, Palmer O, Bartos J, Wang X, Robbins RC, Fischbein MP. Prevention of transplant coronary artery disease by prenylation inhibitors. J Heart Lung Transplant 2011; 30:761-9. [PMID: 21458297 DOI: 10.1016/j.healun.2011.01.720] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 01/10/2011] [Accepted: 01/29/2011] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND In this study we systematically dissect the prenylation pathway to better define the mechanism behind statin inhibition in chronic allograft rejection in heart transplants, or transplant coronary artery disease (TCAD). METHODS Utilizing a murine heterotopic heart transplant model, animals received daily treatments of either statin or selective isoprenoid blockade inhibitors to block the four major downstream branches of the mevalonate pathway. TCAD was assessed by morphometric analysis at Day 52. Graft-infiltrating cells, cytokine production, smooth muscle cell proliferation and migration and endothelial cell MHC II expression were detected on Day 7. RESULTS Atorvastatin and two prenylation inhibitors, NE-10790 and manumycin A, significantly reduced TCAD lesions compared with untreated animals. Perillyl alcohol treatment resulted in a trend toward decreased luminal narrowing. Finally, zaragozic acid (cholesterol blockade only) did not alter TCAD severity. Statins and prenylation inhibitors reduced inflammatory cell allograft recruitment, but did not always correlate with TCAD reduction. Cytokine production was decreased in recipient spleens in all treatment groups. Both in vitro and in vivo IFN-γ-stimulated MHC II expression was decreased in a dose-dependent manner in the atorvastatin, perillyl alcohol and NE-10790 groups. In vitro smooth muscle cell proliferation was decreased in all treatment groups. Finally, in vitro smooth muscle cell migration was decreased in the atorvastatin, NE-10790 and manumycin A groups only. CONCLUSIONS FPT and GGPT-2 (inhibition) are the key enzymes in the HGM-CoA reductase pathway and most influential in TCAD prevention. TCAD reduction is most closely related to smooth muscle cell migration, but not its anti-inflammatory properties.
Collapse
Affiliation(s)
- William Stein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Mitogen-activated protein kinases activation in T lymphocytes of patients with acute coronary syndromes. Basic Res Cardiol 2011; 106:667-79. [DOI: 10.1007/s00395-011-0172-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 02/17/2011] [Accepted: 03/09/2011] [Indexed: 10/18/2022]
|
32
|
Sadowitz B, Maier KG, Gahtan V. Basic Science Review: Statin Therapy-Part I: The Pleiotropic Effects of Statins in Cardiovascular Disease. Vasc Endovascular Surg 2010; 44:241-51. [DOI: 10.1177/1538574410362922] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
3-hydroxy-3-methylglutaryl coenzyme A reductase (HMG CoA-reductase) inhibitors, otherwise known as statins, are currently the medical treatment of choice for hypercholesterolemia. Hypercholesterolemia is a known risk factor for cardiovascular disease, and statin therapy has led to a significant reduction in morbidity and mortality from adverse cardiac events, stroke, and peripheral arterial disease. In addition to achieving a therapeutic decrease in serum cholesterol levels, statin therapy appears to promote other effects that are independent of changes in serum cholesterol. These ‘‘pleiotropic’’ effects include attenuation of vascular inflammation, improved endothelial cell function, stabilization of atherosclerotic plaque, decreased vascular smooth muscle cell migration and proliferation, and inhibition of platelet aggregation. This article is part I of a 2-part review, and it focuses on the pleiotropic effects of statins at the cellular level.
Collapse
Affiliation(s)
- Benjamin Sadowitz
- SUNY Upstate Medical University, Division of Vascular Surgery and Endovascular Services, Syracuse, NY, USA, Department of Veterans Affairs VA Healthcare Network Upstate New York at Syracuse, Syracuse, NY, USA
| | - Kristopher G. Maier
- SUNY Upstate Medical University, Division of Vascular Surgery and Endovascular Services, Syracuse, NY, USA, Department of Veterans Affairs VA Healthcare Network Upstate New York at Syracuse, Syracuse, NY, USA,
| | - Vivian Gahtan
- SUNY Upstate Medical University, Division of Vascular Surgery and Endovascular Services, Syracuse, NY, USA, Department of Veterans Affairs VA Healthcare Network Upstate New York at Syracuse, Syracuse, NY, USA
| |
Collapse
|
33
|
Elia L, Quintavalle M, Zhang J, Contu R, Cossu L, Latronico MVG, Peterson KL, Indolfi C, Catalucci D, Chen J, Courtneidge SA, Condorelli G. The knockout of miR-143 and -145 alters smooth muscle cell maintenance and vascular homeostasis in mice: correlates with human disease. Cell Death Differ 2009; 16:1590-8. [PMID: 19816508 PMCID: PMC3014107 DOI: 10.1038/cdd.2009.153] [Citation(s) in RCA: 445] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mechanisms controlling vascular smooth muscle cell (VSMC) plasticity and renewal still remain to be elucidated completely. A class of small RNAs called microRNAs (miRs) regulate gene expression at the post-transcriptional level. Here, we show a critical role of the miR-143/145 cluster in SMC differentiation and vascular pathogenesis, also through the generation of a mouse model of miR-143 and -145 knockout (KO). We determined that the expression of miR-143 and -145 is decreased in acute and chronic vascular stress (transverse aortic constriction and in aortas of the ApoE KO mouse). In human aortic aneurysms, the expression of miR-143 and -145 was significantly decreased compared with control aortas. In addition, overexpression of miR-143 and -145 decreased neointimal formation in a rat model of acute vascular injury. An in-depth analysis of the miR-143/145 KO mouse model showed that this miR cluster is expressed mostly in the SMC compartment, both during development and postnatally, in vessels and SMC-containing organs. Loss of miR-143 and miR-145 expression induces structural modifications of the aorta, because of an incomplete differentiation of VSMCs. In conclusion, our results show that the miR-143/145 gene cluster has a critical role during SMC differentiation and strongly suggest its involvement in the reversion of the VSMC differentiation phenotype that occurs during vascular disease.
Collapse
Affiliation(s)
- Leonardo Elia
- Department of Medicine, Division of Cardiology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0613C, USA
- Institute of Biomedical Technologies, Consiglio Nazionale delle Ricerche, via Fratelli Cervi 93, 20138 Milan, Italy
| | - Manuela Quintavalle
- Burnham Institute for Medical Research 10901 North Torrey Pines Road, La Jolla, CA 92037
| | - Jianlin Zhang
- Department of Medicine, Division of Cardiology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0613C, USA
| | - Riccardo Contu
- I.R.C.C.S. MultiMedica Hospital, via Gaudenzio Fantoli 16/15, 20138 Milan, Italy
| | - Luca Cossu
- I.R.C.C.S. MultiMedica Hospital, via Gaudenzio Fantoli 16/15, 20138 Milan, Italy
| | | | - Kirk L. Peterson
- Department of Medicine, Division of Cardiology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0613C, USA
| | - Ciro Indolfi
- University Magna Graecia, 88100 Catanzaro, Italy
| | - Daniele Catalucci
- Institute of Biomedical Technologies, Consiglio Nazionale delle Ricerche, via Fratelli Cervi 93, 20138 Milan, Italy
- I.R.C.C.S. MultiMedica Hospital, via Gaudenzio Fantoli 16/15, 20138 Milan, Italy
| | - Ju Chen
- Department of Medicine, Division of Cardiology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0613C, USA
| | - Sara A. Courtneidge
- Burnham Institute for Medical Research 10901 North Torrey Pines Road, La Jolla, CA 92037
| | - Gianluigi Condorelli
- Department of Medicine, Division of Cardiology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0613C, USA
- Institute of Biomedical Technologies, Consiglio Nazionale delle Ricerche, via Fratelli Cervi 93, 20138 Milan, Italy
- I.R.C.C.S. MultiMedica Hospital, via Gaudenzio Fantoli 16/15, 20138 Milan, Italy
| |
Collapse
|
34
|
Torella D, Gasparri C, Ellison GM, Curcio A, Leone A, Vicinanza C, Galuppo V, Mendicino I, Sacco W, Aquila I, Surace FC, Luposella M, Stillo G, Agosti V, Cosentino C, Avvedimento EV, Indolfi C. Differential regulation of vascular smooth muscle and endothelial cell proliferation in vitro and in vivo by cAMP/PKA-activated p85alphaPI3K. Am J Physiol Heart Circ Physiol 2009; 297:H2015-25. [PMID: 19783773 DOI: 10.1152/ajpheart.00738.2009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
cAMP inhibits proliferation in most cell types, triggering different and sometimes opposing molecular pathways. p85alpha (phosphatidylinositol 3-kinase regulatory subunit) is phosphorylated by cAMP/PKA in certain cell lineages, but its effects on vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) are unknown. In the present study, we evaluated 1) the role of p85alpha in the integration of cAMP/PKA-dependent signaling on the regulation of VSMC and EC growth in vitro; and 2) the effects of PKA-modified p85alpha on neointimal hyperplasia and endothelial healing after balloon injury in vivo. Plasmid constructs carrying wild-type and PKA-modified p85alpha were employed in VSMCs and ECs in vitro and after balloon injury in rat carotid arteries in vivo. cAMP/PKA reduced VSMC proliferation through p85alpha phosphorylation. Transfected PKA-activated p85alpha binds p21ras, reducing ERK1/2 activation and VSMC proliferation in vitro. In contrast, EC proliferation inhibition by cAMP is independent from PKA modification of p85alpha and ERK1/2 inhibition; indeed, PKA-activated p85alpha did not inhibit per se ERK1/2 activation and proliferation in ECs in vitro. Interestingly, cAMP reduced both VSMC and EC apoptotic death through p85alpha phosphorylation. Accordingly, PKA-activated p85alpha triggered Akt activation, reducing both VSMC and EC apoptosis in vitro. Finally, compared with controls, vascular gene transfer of PKA-activated p85alpha significantly reduced neointimal formation after balloon injury in rats, without inhibiting endothelial regeneration of the injured arterial segment. In conclusions, PKA-activated p85alpha integrates cAMP/PKA signaling differently in VSMCs and ECs. By reducing neointimal hyperplasia without inhibiting endothelial regeneration, it exerts a protective effect against restenosis after balloon injury.
Collapse
Affiliation(s)
- Daniele Torella
- Laboratory of Molecular and Cellular Cardiology, Magna Graecia University, Campus S. Venuta, Viale Europa-Germaneto, Catanzaro 88100, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Bolchi C, Pallavicini M, Fumagalli L, Ferri N, Corsini A, Rusconi C, Valoti E. New Ras CAAX mimetics: design, synthesis, antiproliferative activity, and RAS prenylation inhibition. Bioorg Med Chem Lett 2009; 19:5500-4. [PMID: 19666221 DOI: 10.1016/j.bmcl.2009.07.065] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2009] [Revised: 07/09/2009] [Accepted: 07/10/2009] [Indexed: 10/20/2022]
Abstract
Mimetics of the C-terminal CAAX tetrapeptide of Ras protein were designed replacing cysteine (C) by 2-hydroxymethylbenzodioxane or 2-aminomethylbenzodioxane, respectively etherified and amidified with 2'-methyl or 2'-methoxy substituted 2-carboxy-4-hydroxybiphenyl and 2,4-dicarboxybiphenyl. These pluri-substituted biphenyl systems, used as internal spacer and AA dipeptide bioisoster, were linked to the methyl ester of l-methionine, glycine or l-leucine by an amide bond. The resultant twelve pairs of stereoisomers at the dioxane C-2 were tested for antiproliferative effect finding the maximum activity for derivatives with methyleneoxy linker between benzodioxane and 2'-methylbiphenyl. Of these compounds, the one with terminal methionine and S configuration proved a good Ras prenylation inhibitor in a cell-based assay.
Collapse
Affiliation(s)
- Cristiano Bolchi
- Dipartimento di Scienze Farmaceutiche 'Pietro Pratesi', Università degli Studi di Milano, via Mangiagalli 25, I-20133 Milano, Italy
| | | | | | | | | | | | | |
Collapse
|
36
|
Ramos KS. H-RAS controls phenotypic profiles of vascular smooth muscle cells and the pathogenesis of vascular proliferative disorders. Circ Res 2009; 104:1139-41. [PMID: 19461105 DOI: 10.1161/circresaha.109.199554] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
37
|
Sedding DG, Tröbs M, Reich F, Walker G, Fink L, Haberbosch W, Rau W, Tillmanns H, Preissner KT, Bohle RM, Langheinrich AC. 3-Deazaadenosine prevents smooth muscle cell proliferation and neointima formation by interfering with Ras signaling. Circ Res 2009; 104:1192-200. [PMID: 19372464 DOI: 10.1161/circresaha.109.194357] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
3-Deazaadenosine (c3Ado) is a potent inhibitor of S-adenosylhomocysteine hydrolase, which regulates cellular methyltransferase activity. In the present study, we sought to determine the effect of c3Ado on vascular smooth muscle cell (VSMC) function and neointima formation in vivo. c3Ado dose-dependently prevented the proliferation and migration of human coronary VSMCs in vitro. This was accompanied by an increased expression of the cyclin-dependent kinase inhibitors p21(WAF1/Cip1), p27(Kip1), a decreased expression of G(1)/S phase cyclins, and a lack of retinoblastoma protein hyperphosphorylation. In accordance with these findings, fluorescence-activated cell-sorting analysis of propidium iodide-stained cells indicated a cell cycle arrest in the G(0)/G(1) phase. Importantly, c3Ado did not affect the number of viable (trypan blue exclusion) or apoptotic cells (TUNEL). Mechanistically, c3Ado prevented FCS-induced Ras carboxyl methylation and membrane translocation and activity by inhibiting isoprenylcysteine carboxyl methyltransferase and reduced FCS-induced extracellular signal-regulated kinase (ERK)1/2 and Akt phosphorylation in a dose-dependent manner. Conversely, rescuing signal transduction by overexpression of a constitutive active Ras mutant abrogated c3Ado's effect on proliferation. For in vivo studies, the femoral artery of C57BL/6 mice was dilated and mice were fed a diet containing 150 microg of c3Ado per day. c3Ado prevented dilation-induced Ras activation, as well as ERK1/2 and Akt phosphorylation in vivo. At day 21, VSMC proliferation (proliferating-cell nuclear antigen [PCNA]-positive cells), as well as the neointima/media ratio (0.7+/-0.2 versus 1.6+/-0.4; P<0.05) were significantly reduced, without any changes in the number of apoptotic cells. Our data indicate that c3Ado interferes with Ras methylation and function and thereby with mitogenic activation of ERK1/2 and Akt, preventing VSMC cell cycle entry and proliferation and neointima formation in vivo. Thus, therapeutic inhibition of S-adenosylhomocysteine hydrolase by c3Ado may represent a save and effective novel approach to prevent vascular proliferative disease.
Collapse
Affiliation(s)
- Daniel G Sedding
- Department of Internal Medicine I/Cardiology, Giessen University, Klinikstrasse 36, 35392 Giessen, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Coats P, Kennedy S, Pyne S, Wainwright CL, Wadsworth RM. Inhibition of non-Ras protein farnesylation reduces in-stent restenosis. Atherosclerosis 2008; 197:515-23. [PMID: 17662987 DOI: 10.1016/j.atherosclerosis.2007.06.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2007] [Revised: 06/11/2007] [Accepted: 06/19/2007] [Indexed: 11/27/2022]
Abstract
Ras has a key role in relation to cell proliferation, survival and migration and requires farnesylation for full activity. The effects of a Ras farnesyl transferase inhibitor, FPT III on human atherosclerotic vascular smooth muscle (VSM) cells proliferation and p42/p44 mitogen-activated protein kinase (p42/p44 MAPK) activity was measured. In addition the ability of FPT III to modify the development of neointimal growth was tested in cultured human arteries and in a rabbit model of in-stent restenosis. In human VSM cells FPT III (25 microM) inhibited FCS-stimulated cell proliferation through a ras-dependent mechanism (after 18 h exposure) and also a novel ras-independent mechanism (following 15 min exposure). FPT III incubation (18 h) inhibited platelet-derived growth factor (PDGF)-stimulated p42/p44 MAPK activation and p21 Ras membrane localization, whereas 15 min incubation had no effect on the activation of p42/p44 MAPK in response to PDGF (added at 18 h) or on membrane p21 Ras localization (measured at 18 h). In cultured human atherosclerotic arteries, the presence of 25 microM FPT III significantly reduced neointimal growth. In vivo, 15 min local infusion of 25 microM FPT III significantly reduced in-stent restenosis 28 days later without affecting vascular function in normal rabbit artery. This study demonstrates that brief administration of a farnesyl transferase inhibitor reduced in-stent restenosis in a rabbit model without deleterious effects on vascular function or endothelial regrowth. Acute application of FPT III was found to act through a novel mechanism to inhibit smooth muscle cell proliferation via a non-ras pathway, which may contribute to the prevention of in-stent restenosis.
Collapse
Affiliation(s)
- Paul Coats
- Division of Physiology and Pharmacology, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 27 Taylor Street, Glasgow G4 0NR, Scotland, UK
| | | | | | | | | |
Collapse
|
39
|
Denèfle P, Duverger N, Branellec D. Section Review: Cardiovascular & Renal: Genetic therapies for vascular diseases and lipid disorders. Expert Opin Investig Drugs 2008. [DOI: 10.1517/13543784.4.11.1129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
40
|
Guo X, Chen KH, Guo Y, Liao H, Tang J, Xiao RP. Mitofusin 2 Triggers Vascular Smooth Muscle Cell Apoptosis via Mitochondrial Death Pathway. Circ Res 2007; 101:1113-22. [PMID: 17901359 DOI: 10.1161/circresaha.107.157644] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Previous studies have shown that mitofusin 2 (Mfn-2) (or hyperplasia suppressor gene [HSG]) inhibits vascular smooth muscle cell (VSMC) proliferation. Here, we demonstrate that Mfn-2 is a primary determinant of VSMC apoptosis. First, oxidative stress with H2O2, inhibition of protein kinase C with staurosporine, activation of protein kinase A with forskolin, and serum deprivation concurrently elevate Mfn-2 expression and induce VSMC apoptosis. Second, overexpression of Mfn-2 also triggers apoptosis of VSMCs in culture and in balloon-injured rat carotid arteries, thus contributing to Mfn-2-mediated prevention of neointima formation after angioplasty. Third, Mfn-2 silencing protects VSMCs against H2O2 or Mfn-2 overexpression-induced apoptosis, indicating that upregulation of Mfn-2 is necessary and sufficient for oxidative stress-mediated VSMC apoptosis. The Mfn-2 proapoptotic effect is independent of its role in mitochondrial fusion but mainly mediated by inhibition of Akt signaling and the resultant activation of the mitochondrial apoptotic pathway, as manifested by decreased Akt phosphorylation, increased mitochondrial Bax/Bcl-2 ratio, cytochrome c release, and activation of caspases-9 and caspase-3. Furthermore, Mfn-2-induced apoptosis was blocked by overexpression of an active phosphoinositide 3-kinase mutant or Bcl-xL or inhibition of caspase-9 but not caspases-8. Thus, in addition to its antiproliferative effects, Mfn-2 constitutes a primary determinant of VSMC apoptosis.
Collapse
Affiliation(s)
- Xiaomei Guo
- Laboratory of Cardiovascular Science, Gerontology Research Center, NIA, NIH, 5600 Nathan Shock Dr, Baltimore, MD 21224, USA
| | | | | | | | | | | |
Collapse
|
41
|
Bolchi C, Pallavicini M, Rusconi C, Diomede L, Ferri N, Corsini A, Fumagalli L, Pedretti A, Vistoli G, Valoti E. Peptidomimetic inhibitors of farnesyltransferase with high in vitro activity and significant cellular potency. Bioorg Med Chem Lett 2007; 17:6192-6. [PMID: 17889533 DOI: 10.1016/j.bmcl.2007.09.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Revised: 09/05/2007] [Accepted: 09/05/2007] [Indexed: 11/22/2022]
Abstract
2-o-Tolyl or 2-o-anisyl substituted 4-hydroxy- and 4-carboxybenzamides of methionine, etherified and amidified with 2-hydroxymethyl- and 2-aminomethylpyridodioxane, respectively, are described as inhibitors of Ras protein farnesyltransferase (FTase). Of the sixteen compounds, resulting from the substitution pattern of benzamide and the configuration of the two stereocenters, seven inhibited FTase activity with potencies in the nanomolar range. They were all 2-oxymethylpyridodioxane ethers and, among them, the four o-tolyl substituted stereoisomers also showed micromolar antiproliferative effect on human aortic smooth muscle cells interfering with Ras farnesylation. The docking analysis enlightened significant differences in enzyme interaction between oxymethylpyridodioxane and aminomethylpyridodioxane derivatives.
Collapse
Affiliation(s)
- Cristiano Bolchi
- Istituto di Chimica Farmaceutica e Tossicologica Pietro Pratesi, Università di Milano, via Mangiagalli 25, Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
De Flora S, Izzotti A. Mutagenesis and cardiovascular diseases Molecular mechanisms, risk factors, and protective factors. Mutat Res 2007; 621:5-17. [PMID: 17383689 DOI: 10.1016/j.mrfmmm.2006.12.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Revised: 12/12/2006] [Accepted: 12/13/2006] [Indexed: 05/14/2023]
Abstract
Although no generalization can be made, it is of interest that cancer, cardiovascular diseases, and other chronic conditions often share common risk factors and common protective factors as well as common pathogenetic determinants, such as DNA damage, oxidative stress, and chronic inflammation. Atherosclerosis is the most important cause of vascular forms representing the major cause of death in the population of many geographical areas. A great deal of studies support the "response-to-injury" theory. A variety of experimental and epidemiological findings are also in favor of the somatic mutation theory, which maintains that the earliest event in the atherogenic process is represented by mutations in arterial smooth muscle cells, akin to formation of a benign tumor. These two theories can be harmonized, also taking into account the highly diversified nature of atherosclerotic lesions. Molecular epidemiology studies performed in our laboratory and other laboratories have shown that DNA adducts are systematically present in arterial smooth muscle cells, and their levels are correlated with atherogenic risk factors known from traditional epidemiology. Oxidative DNA damage was also consistently detected in these cells. The role of glutathione S-transferase polymorphisms on the frequency of the above molecular alterations and of arterial diseases is rather controversial. Prevention of both cancer and atherosclerosis is based on avoidance of exposure to risk factors and on fortification of the host defense mechanisms by means of dietary principles and chemopreventive drugs.
Collapse
Affiliation(s)
- Silvio De Flora
- Department of Health Sciences, University of Genoa, Via A. Pastore 1, I-16132 Genoa, Italy.
| | | |
Collapse
|
43
|
Torella D, Curcio A, Gasparri C, Galuppo V, De Serio D, Surace FC, Cavaliere AL, Leone A, Coppola C, Ellison GM, Indolfi C. Fludarabine prevents smooth muscle proliferation in vitro and neointimal hyperplasia in vivo through specific inhibition of STAT-1 activation. Am J Physiol Heart Circ Physiol 2007; 292:H2935-43. [PMID: 17293493 DOI: 10.1152/ajpheart.00887.2006] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Drug-eluting stents are increasingly used to reduce in-stent restenosis and adverse cardiac events after percutaneous coronary interventions. However, the race for the ideal drug-eluting stent is still on, with special regard to the best stent-coating system and the most effective and less toxic drug. Fludarabine, a nucleoside analog, has both anti-inflammatory and antiproliferative cellular effects. The aim of the present study was to assess the cellular and molecular effects of fludarabine on vascular smooth muscle cell (VSMC) growth in vitro and in vivo and the feasibility and efficacy of a fludarabine-eluting stent. To study the biomolecular effects of fludarabine on VSMC proliferation in vitro, rat VSMCs were grown in the presence of 50 μM fludarabine or in the absence of the same. To evaluate the in vivo effect of this drug, male Wistar rats underwent balloon injury of the carotid artery, and fludarabine was locally delivered at the time of injury. Finally, fludarabine-eluting stents were in-laboratory manufactured and tested in a rabbit model of in-stent restenosis. Fludarabine markedly inhibited VSMC proliferation in cell culture. Furthermore, fludarabine reduced neointimal formation after balloon angioplasty in a dose-dependent manner, and fludarabine-eluting stents reduced neointimal hyperplasia by ∼50%. These in vitro and in vivo cellular effects were specifically associated with the molecular switch-off of signal transducer and activator of transcription (STAT)-1 activation, without affecting other STAT proteins. Fludarabine abolishes VSMC proliferation in vitro and reduces neointimal formation after balloon injury in vivo through specific inhibition of STAT-1 activation. Fludarabine-eluting stents are feasible and effective in reducing in-stent restenosis in rabbits.
Collapse
MESH Headings
- Angioplasty, Balloon/adverse effects
- Animals
- Aorta/drug effects
- Aorta/metabolism
- Cardiovascular Agents/administration & dosage
- Cardiovascular Agents/pharmacology
- Cardiovascular Agents/therapeutic use
- Carotid Artery Injuries/etiology
- Carotid Artery Injuries/pathology
- Carotid Artery Injuries/prevention & control
- Carotid Stenosis/etiology
- Carotid Stenosis/pathology
- Carotid Stenosis/prevention & control
- Cell Proliferation/drug effects
- Cells, Cultured
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Feasibility Studies
- Hyperplasia
- Janus Kinase 2/metabolism
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Phosphorylation
- Prosthesis Design
- RNA, Antisense/genetics
- RNA, Antisense/metabolism
- Rabbits
- Rats
- Rats, Wistar
- STAT1 Transcription Factor/genetics
- STAT1 Transcription Factor/metabolism
- Stents/adverse effects
- Time Factors
- Transfection
- Tunica Intima/drug effects
- Tunica Intima/pathology
- Vidarabine/administration & dosage
- Vidarabine/analogs & derivatives
- Vidarabine/pharmacology
- Vidarabine/therapeutic use
Collapse
Affiliation(s)
- Daniele Torella
- Laboratory of Molecular and Cellular Cardiology, Magna Graecia University, Catanzaro, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Sugita M, Sugita H, Kaneki M. Farnesyltransferase Inhibitor, Manumycin A, Prevents Atherosclerosis Development and Reduces Oxidative Stress in Apolipoprotein E-Deficient Mice. Arterioscler Thromb Vasc Biol 2007; 27:1390-5. [PMID: 17363690 DOI: 10.1161/atvbaha.107.140673] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Statins are presumed to exert their antiatherogenic effects in part via lipid-lowering-independent mechanisms. Inhibition of protein farnesylation and/or geranylgeranylation by statins has been postulated to contribute to the lipid-lowering-independent effects. However, a role for protein farnesylation in atherogenesis has not yet been studied. Therefore, we examined the effects of farnesyltransferase inhibitor, manumycin A, on the development of atherosclerosis in apolipoprotein E (apoE)-deficient mice fed a high-fat diet. METHODS AND RESULTS Manumycin A treatment for 22 weeks decreased Ras activity, and reduced fatty streak lesion size at the aortic sinus to 43% of that in vehicle-treated apoE-deficient mice (P<0.05), while plasma total cholesterol was unaltered. Moreover, manumycin A reduced alpha-smooth muscle actin-positive area to 29% of that in vehicle-treated apoE-deficient mice (P<0.01). The prevention of atherogenesis by manumycin A was accompanied by amelioration of oxidative stress, as judged by reduced ex vivo superoxide production and nitrotyrosine immunoreactivity. CONCLUSIONS These results indicate that the inhibition of farnesyltransferase prevents the development of mature atherosclerosis with concomitant alleviation of oxidative stress in apoE-deficient mice. The present data highlight farnesyltransferase as a potential molecular target for preventive and/or therapeutic intervention against atherosclerosis.
Collapse
Affiliation(s)
- Michiko Sugita
- Department of Anesthesia & Critical Care, Massachusetts General Hospital, Harvard Medical School, 149 Thirteenth Street, Rm. 6604, Charlestown, MA 02129, USA
| | | | | |
Collapse
|
45
|
Abstract
The introduction of percutaneous transluminal coronary angioplasty has revolutionized the field of cardiology by providing patients with coronary artery disease immediate and effective therapy. Overshadowing the early success of angioplasty was the high rate of angiographic restenosis and recurrent symptoms at 6 months. The use of stents reduced the incidence of restenosis; however, the rise in the number of patients undergoing percutaneous interventions produced a new problem of restenosis occurring within the stent: in-stent restenosis (ISR). Mechanical approaches, including directional and rotational atherectomy and systemic pharmacotherapy, have failed to demonstrate a reduction in ISR in randomized clinical trials. Intravascular brachytherapy is currently the only approved therapy for ISR, although this treatment has numerous unresolved questions and is not effective in a large percent of patients. Drug-eluting stents have reduced the incidence of restenosis by providing localized therapy to the targeted lesion without systemic toxicity. The purpose of this review is to synthesize data from major clinical trials involving the 2 most successful agents used in the prevention of restenosis: sirolimus and paclitaxel. The cellular and molecular mechanisms of both ISR and restenosis postangioplasty derived from animal models will be introduced. Second, an overview of 3 alternate interventions that attempt to reduce the rates of restenosis is presented. Finally, the major randomized, controlled trials involving sirolimus and paclitaxel are described, and their clinical implications and use as a possible solution in the prevention of restenosis is discussed.
Collapse
Affiliation(s)
- Leo Slavin
- Department of Medicine, University of California, Los Angeles, California, USA
| | | | | |
Collapse
|
46
|
Kim J, Keys JR, Eckhart AD. Vascular smooth muscle migration and proliferation in response to lysophosphatidic acid (LPA) is mediated by LPA receptors coupling to Gq. Cell Signal 2006; 18:1695-701. [PMID: 16504475 DOI: 10.1016/j.cellsig.2006.01.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2005] [Revised: 01/12/2006] [Accepted: 01/16/2006] [Indexed: 12/21/2022]
Abstract
Many G protein-coupled receptors can couple to multiple G proteins to convey their intracellular signaling cascades. The receptors for lysophosphatidic acid (LPA) possess this ability. LPA receptors are important mediators of a wide variety of biological actions including cell migration, proliferation and survival which are processes that can all have a considerable impact on vascular smooth muscle (VSM) and blood vessels. To date, confirmation of G proteins involved has mostly relied on the inhibition of Gi-mediated signaling via pertussis toxin (PTx). We were interested in the specific involvement of LPA-Gq-mediated signaling therefore we isolated aorta VSM cells (VSMCs) from transgenic mice that express a peptide inhibitor of Gq, GqI, exclusively in VSM. We detected both LPA1 and LPA2 receptor expression in mouse VSM whereas LPA1 and LPA3 were expressed in rat VSM. SM22-GqI did not alter LPA-induced migration but it was sufficient to attenuate LPA-induced proliferation. GqI expression also attenuated LPA-induced ERK1/2 and Akt activation by 40-50%. To test the feasibility of this peptide as a potential therapeutic agent, we also generated adenovirus encoding the GqI. Transient expression of GqI was capable of inhibiting both LPA-induced migration and proliferation of VSMCs isolated from rat and mouse. Furthermore, ERK activation in response to LPA was also attenuated in VSMCs with Adv-GqI. Therefore, LPA receptors couple to Gq in VSMC and mediate migration and proliferation which may be mediated through activation of ERK1/2 and Akt. Our data also suggest that both chronic and transient expression of the GqI peptide is an effective strategy to lower Gq-mediated LPA signaling and may be a successful therapeutic strategy to combat diseases with enhanced VSM growth such as occurs following angioplasty or stent implantation.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- DNA/biosynthesis
- Enzyme Activation/drug effects
- Extracellular Signal-Regulated MAP Kinases/metabolism
- GTP-Binding Protein alpha Subunits, Gq-G11/metabolism
- Gene Expression Regulation/drug effects
- Lysophospholipids/pharmacology
- Mice
- Mice, Transgenic
- Microfilament Proteins/metabolism
- Muscle Proteins/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Receptors, Lysophosphatidic Acid/genetics
- Receptors, Lysophosphatidic Acid/metabolism
Collapse
Affiliation(s)
- Jihee Kim
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | |
Collapse
|
47
|
Brewster L, Brey E, Greisler H. Cardiovascular gene delivery: The good road is awaiting. Adv Drug Deliv Rev 2006; 58:604-29. [PMID: 16769148 PMCID: PMC3337725 DOI: 10.1016/j.addr.2006.03.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2005] [Accepted: 03/24/2006] [Indexed: 01/13/2023]
Abstract
Atherosclerotic cardiovascular disease is a leading cause of death worldwide. Despite recent improvements in medical, operative, and endovascular treatments, the number of interventions performed annually continues to increase. Unfortunately, the durability of these interventions is limited acutely by thrombotic complications and later by myointimal hyperplasia followed by progression of atherosclerotic disease over time. Despite improving medical management of patients with atherosclerotic disease, these complications appear to be persisting. Cardiovascular gene therapy has the potential to make significant clinical inroads to limit these complications. This article will review the technical aspects of cardiovascular gene therapy; its application for promoting a functional endothelium, smooth muscle cell growth inhibition, therapeutic angiogenesis, tissue engineered vascular conduits, and discuss the current status of various applicable clinical trials.
Collapse
Affiliation(s)
- L.P. Brewster
- Department of Surgery, Loyola University Medical Center, Maywood, IL, 60153, USA
- Department of Cell Biology, Neurobiology, and Anatomy, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - E.M. Brey
- Department of Surgery, Loyola University Medical Center, Maywood, IL, 60153, USA
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, 60616, USA
- Research and Surgical Services, Edward J. Hines Jr. V.A. Hospital, Hines, IL, 60141, USA
| | - H.P. Greisler
- Department of Surgery, Loyola University Medical Center, Maywood, IL, 60153, USA
- Department of Cell Biology, Neurobiology, and Anatomy, Loyola University Medical Center, Maywood, IL, 60153, USA
- Research and Surgical Services, Edward J. Hines Jr. V.A. Hospital, Hines, IL, 60141, USA
- Corresponding author. Loyola University Medical Center, Department of Surgery, 2160 South First Avenue, Maywood, IL, 60153, USA. Tel.: +1 708 216 8541; fax: +1 708 216 6300. (H.P. Greisler)
| |
Collapse
|
48
|
Patterson C, Mapera S, Li HH, Madamanchi N, Hilliard E, Lineberger R, Herrmann R, Charles P. Comparative Effects of Paclitaxel and Rapamycin on Smooth Muscle Migration and Survival. Arterioscler Thromb Vasc Biol 2006; 26:1473-80. [PMID: 16645158 DOI: 10.1161/01.atv.0000223866.42883.3b] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Advances in stent technology have enabled the delivery of drugs to improve outcomes after stent deployment. However, the optimal payloads for stents are not clear, and the appropriate stent-based therapies for high-risk patients, such as diabetics, have not been clearly established.
Methods and Results—
We used smooth muscle cell culture models to compare the activities of rapamycin and paclitaxel. Smooth muscle cells were grown in normal or high glucose to induce insulin resistance. Both paclitaxel and rapamycin activate mitogen-activated protein kinase pathways similarly. However, rapamycin potently activates AKT-dependent signaling, an effect that overrides the downregulation of this pathway by insulin resistance and that causes phosphorylation of the AKT-dependent transcription factor FOXO1. This effect is associated with attenuation of the anti-migratory effects of rapamycin under high glucose conditions that are not observed with paclitaxel, as well as with increased protection against ceramide-induced cytotoxicity, both of which are dependent on FOXO1 phosphorylation.
Conclusions—
Differences between the ability of rapamycin and paclitaxel to activate AKT may account for their differential cell survival and antichemotactic activities. These observations may provide a basis for understanding clinical differences between rapamycin- and paclitaxel-coated stents. The approaches used in these studies can be expanded to other candidate stent payloads as a method for triage in preclinical studies.
Collapse
Affiliation(s)
- Cam Patterson
- Division of Cardiology and Carolina Cardiovascular Biology Center, University of North Carolina at Chapel Hill, 8200 Medical Biomolecular Research Building, Chapel Hill, NC 27599-7126, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Schiele TM. Current understanding of coronary in-stent restenosis. Pathophysiology, clinical presentation, diagnostic work-up, and management. ACTA ACUST UNITED AC 2006; 94:772-90. [PMID: 16258781 DOI: 10.1007/s00392-005-0299-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2005] [Accepted: 07/18/2005] [Indexed: 12/29/2022]
Abstract
In-stent restenosis is the limiting entity following coronary stent implantation. It is associated with significant morbidity and cost and thus represents a major clinical and economical problem. Worldwide, approximately 250 000 in-stent restenotic lesions per year have to be dealt with. The pathophysiology of instent restenosis is multifactorial and comprises inflammation, smooth muscle cell migration and proliferation and extracellular matrix formation, all mediated by distinct molecular pathways. Instent restenosis has been recognised as very difficult to manage, with a repeat restenosis rate of 50% regardless of the mechanical angioplasty device used. Much more favourable results were reported for the adjunctive irradiation of the in-stent restenotic lesion, with a consistent reduction of the incidence of repeat in-stent restenosis by 50%. Data from the first clinical trials on drug-eluting stents for the treatment of in-stent restenosis have shown very much promise yielding this strategy likely to become the treatment of choice. This review outlines the histological and molecular findings of the pathophysiology, the epidemiology, the predictors and the diagnostic work-up of in-stent restenosis and puts emphasis on the various treatment options for its prevention and therapy.
Collapse
Affiliation(s)
- T M Schiele
- Kardiologie, Klinikum der Ludwig-Maximilians-Universität München--Innenstadt, Ziemssenstrasse 1, 80336 München, Germany.
| |
Collapse
|
50
|
Fields RC, Baig K, Gaca J, Milton LG, Koch WJ, Lawson JH. Reduction of vascular intimal-medial hyperplasia in polytetrafluoroethylene arteriovenous grafts via expression of an inhibitor of G protein signaling. Ann Vasc Surg 2005; 19:712-8. [PMID: 16075345 DOI: 10.1007/s10016-005-6805-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Polytetrafluoroethylene (PTFE) arteriovenous (AV) grafts are performed routinely for vascular access. The limited life span of PTFE grafts is a major cause of morbidity. Graft failure is attributed to venous outflow tract vascular smooth muscle (VSM) hyperplasia, which is linked to heterotrimeric G protein signaling. We proposed that expression of a peptide inhibitor of G(betagamma) signaling (betaARKct) in the venous outflow of PTFE grafts would reduce hyperplasia and prolong graft patency. Left carotid to right external jugular vein PTFE AV grafts were placed in swine. The isolated external jugular vein was treated with an adenovirus encoding betaARKct, empty adenovirus, or phosphate-buffered saline for approximately 25 min. After 7 or 28 days, flow probe analysis was performed and the vein was harvested and analyzed for cross-sectional area comparison. After both 7 and 28 days, when compared to controls, treated animals demonstrated a statistically significant reduction in VSM hyperplasia with a reduction in cross-sectional intimal and medial areas of >40% (p < 0.05). Flow was maintained in treated grafts, while control groups demonstrated a >50% reduction (p < 0.05) at 7 days. Further, treated grafts demonstrated significant improvement in graft patency at 28 days (100% vs. 12% for treated and untreated grafts, respectively). The inhibition of G(betagamma) signaling reduces intimal-medial hyperplasia and prolongs graft patency in PTFE AV grafts. This represents a novel molecular therapeutic strategy for improving the patency of vascular access grafts.
Collapse
Affiliation(s)
- Ryan C Fields
- Department of Surgery, Duke University Medical Center, DUMC Box 2622, Room 481 MSRB, Research Drive, Durham, NC, 27710, USA
| | | | | | | | | | | |
Collapse
|