1
|
Jing X, Fu H, Yu B, Sun M, Wang L. Two-photon polymerization for 3D biomedical scaffolds: Overview and updates. Front Bioeng Biotechnol 2022; 10:994355. [PMID: 36072288 PMCID: PMC9441635 DOI: 10.3389/fbioe.2022.994355] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 07/29/2022] [Indexed: 01/23/2023] Open
Abstract
The needs for high-resolution, well-defined and complex 3D microstructures in diverse fields call for the rapid development of novel 3D microfabrication techniques. Among those, two-photon polymerization (TPP) attracted extensive attention owing to its unique and useful characteristics. As an approach to implementing additive manufacturing, TPP has truly 3D writing ability to fabricate artificially designed constructs with arbitrary geometry. The spatial resolution of the manufactured structures via TPP can exceed the diffraction limit. The 3D structures fabricated by TPP could properly mimic the microenvironment of natural extracellular matrix, providing powerful tools for the study of cell behavior. TPP can meet the requirements of manufacturing technique for 3D scaffolds (engineering cell culture matrices) used in cytobiology, tissue engineering and regenerative medicine. In this review, we demonstrated the development in 3D microfabrication techniques and we presented an overview of the applications of TPP as an advanced manufacturing technique in complex 3D biomedical scaffolds fabrication. Given this multidisciplinary field, we discussed the perspectives of physics, materials science, chemistry, biomedicine and mechanical engineering. Additionally, we dived into the principles of tow-photon absorption (TPA) and TPP, requirements of 3D biomedical scaffolders, developed-to-date materials and chemical approaches used by TPP and manufacturing strategies based on mechanical engineering. In the end, we draw out the limitations of TPP on 3D manufacturing for now along with some prospects of its future outlook towards the fabrication of 3D biomedical scaffolds.
Collapse
Affiliation(s)
- Xian Jing
- Key Laboratory of Micro/Nano and Ultra-precision Manufacturing, School of Mechatronic Engineering, Changchun University of Technology, Changchun, Jilin, China
| | - Hongxun Fu
- Key Laboratory of Micro/Nano and Ultra-precision Manufacturing, School of Mechatronic Engineering, Changchun University of Technology, Changchun, Jilin, China
| | - Baojun Yu
- Key Laboratory of Micro/Nano and Ultra-precision Manufacturing, School of Mechatronic Engineering, Changchun University of Technology, Changchun, Jilin, China
| | - Meiyan Sun
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Liye Wang
- College of Pharmacy, University of Houston, Houston, TX, United States
| |
Collapse
|
2
|
Zhu R, Liu TW, Liu F. Exogenous Urokinase Inhibits Proteasomal Degradation of Its Cognate Urokinase Plasminogen Activator Receptor. Front Pharmacol 2022; 13:754271. [PMID: 36034808 PMCID: PMC9411529 DOI: 10.3389/fphar.2022.754271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Acute pulmonary embolism (APE) is a debilitating condition with high incidence and mortality rates. APE is widely treated with the serine protease urokinase or urokinase-type plasminogen activator (uPA) that functions by resolving blood clots via catalyzing the conversion of plasminogen to plasmin. Treatment with recombinant uPA has been shown to increase endogenous expression of uPA and its cognate receptor, uPAR; however, the mechanisms for this induction are not known. Using an in vitro hypoxia/reoxygenation model in bronchial epithelial BEAS-2B cells, we show that induction of hypoxia/reoxygenation induces apoptosis and increases secretion of tumor necrosis factor–alpha, brain natriuretic peptide, and fractalkine, which are attenuated when treated with exogenous uPA. Induction of hypoxia/reoxygenation resulted in decreased expression of uPAR on cell surface without any significant changes in its messenger RNA expression, highlighting post-transcriptional regulatory mechanisms. Determination of uPAR protein half-life using cycloheximide showed treatment with uPA significantly increased its half-life (209.6 ± 0.2 min from 48.2 ± 2.3 min). Hypoxia/reoxygenation promoted the degradation of uPAR. Inhibition of proteasome-mediated degradation using MG-132 and lactacystin revealed that uPAR was actively degraded when hypoxia/reoxygenation was induced and that it was reversed when treated with exogenous uPA. Determination of the proteolytic activity of 20S proteasome showed a global increase in ubiquitin–proteasome activation without an increase in proteasome content in cells subjected to hypoxia/reoxygenation. Our results cumulatively reveal that uPAR is actively degraded following hypoxia/reoxygenation, and the degradation was significantly weakened by exogenous uPA treatment. Given the importance of the uPA/uPAR axis in a multitude of pathophysiological contexts, these findings provide important yet undefined mechanistic insights.
Collapse
Affiliation(s)
- Ran Zhu
- Department of Critical Care Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Ting-Wei Liu
- Department of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Fan Liu
- Department of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, China
- *Correspondence: Fan Liu,
| |
Collapse
|
3
|
Dakhel A, Memon AA, Zarrouk M, Ågren-Witteschus S, Sundquist J, Sundquist K, Gottsäter A. Novel cardiovascular biomarkers associated with peripheral arterial disease in men screened for abdominal aortic aneurysm. VASA 2022; 51:167-173. [DOI: 10.1024/0301-1526/a000999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Summary: Background: Peripheral arterial disease (PAD) is a common atherosclerotic disease with severity ranging from asymptomatic to chronic limb threatening ischemia. The aim of the present cross-sectional study was to identify novel biomarkers associated with PAD. Patients and methods: Levels of 91 cardiovascular specific proteins in plasma samples were measured by the Proseek Multiplex CVD III96x96 panel from a cohort consisting of 267 65-year-old men recruited from a screening program for abdominal aortic aneurysm (AAA) Levels of protein biomarkers were compared in men with and without PAD (defined as an ankle brachial index of <0.9) and their diagnostic potential was calculated by receiver-operating characteristic analysis. Results: The prevalence of PAD was 14.2% (38/267). After adjustment for multiple comparisons, levels of the following 11 biomarkers remained significantly higher ( p<0.0001) in patients with PAD: secretoglobin family 3A member 2, osteoprotegerin, urokinase-type plasminogen activator surface receptor, serum macrophage chemokine ligand 16, matrix metalloproteinase 9, p-selectin, growth differentiation factor 15, elafin, cystatin B, trefoil factor 3, and fatty acid-binding protein 4. Multivariable logistic regression analysis (adjusted for smoking, use of antihypertensive and lipid-lowering medication, and metformin) showed that 11 biomarkers were significantly associated with higher risk of PAD with odds ratios ranging from 1.6 to 2.4. Area under curve calculated by receiver operating characteristic curve analysis (diagnostic value) for each protein biomarker ranged from 0.63 to 0.74. Conclusions: We have identified multiple proteins with a potential to be diagnostic biomarkers for PAD, and further research is warranted to clarify their potential predictive and prognostic value.
Collapse
Affiliation(s)
- Ardwan Dakhel
- Department of Vascular Diseases, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Ashfaque A. Memon
- Wallenberg Laboratory, Center for Primary Health Care Research, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Moncef Zarrouk
- Department of Vascular Diseases, Skåne University Hospital, Lund University, Malmö, Sweden
| | | | - Jan Sundquist
- Wallenberg Laboratory, Center for Primary Health Care Research, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Kristina Sundquist
- Wallenberg Laboratory, Center for Primary Health Care Research, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Anders Gottsäter
- Department of Acute and Internal Medicine, Skåne University Hospital, Lund University, Malmö, Sweden
| |
Collapse
|
4
|
Zhu R, Qi WY, Liu TW, Liu F. MicroRNA 449a can Attenuate Protective Effect of Urokinase Against Pulmonary Embolism. Front Pharmacol 2022; 13:713848. [PMID: 35571119 PMCID: PMC9095938 DOI: 10.3389/fphar.2022.713848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 04/08/2022] [Indexed: 11/13/2022] Open
Abstract
Acute pulmonary embolism (APE) is a disabling diseases with high incidence rate and mortality rate. Although with high specificity, D-Dimer lacks specificity to assess APE, hence additional diagnostic and prognostic biomarkers are necessary. APE is widely treated with serine protease urokinase or urokinase-type plasminogen activator (uPA), which act as a catalyst for conversion of plasminogen to plasmin to resolve blood clots. However, it is unknown the role of differential expression of microRNAs (miRNAs) in protective effect of uPA against APE. Hence, we performed miRNA profiling in a hypoxia/reoxygenation (H/R) model of bronchial epithelial BEAS-2B cells in vitro and a APE mice model in vivo. Our analysis revealed that miR-34a-5p, miR-324-5p, miR-331-3p are upregulated with H/R or APE induction, whereas miR-429, miR-491-5p, and miR-449a are downregulated. The differential expression of the miRNAs was attenuated to levels comparable to control by treatment with uPA both in vitro and in vivo. In situ target prediction and analysis of potential functions of the target genes showed that the enrichment of biological processes and pathways were related to cell growth, proliferation, and inflammation. Ectopic overexpression of miR-449a using a mimic completely attenuated the protective effect of uPA in the H/R model in vitro. These results provide a group of miRNAs that could be used as markers, and the modulation of these miRNAs might have potential therapeutic benefits in patients with APE, which need to be validated in additional studies in humans.
Collapse
Affiliation(s)
- Ran Zhu
- Department of Critical Care Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Wei-yi Qi
- Department of Pulmonary and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Ting-wei Liu
- Department of Pulmonary and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Fan Liu
- Department of Pulmonary and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, China
- *Correspondence: Fan Liu,
| |
Collapse
|
5
|
Roles of MicroRNAs in Peripheral Artery In-Stent Restenosis after Endovascular Treatment. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9935671. [PMID: 34368362 PMCID: PMC8337102 DOI: 10.1155/2021/9935671] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 07/13/2021] [Indexed: 12/16/2022]
Abstract
Endovascular repair including percutaneous transluminal angioplasty (PTA) and stent implantation has become the standard approach for the treatment of peripheral arterial disease; however, restenosis is still the main limited complication for the long-term success of the endovascular repair. Endothelial denudation and regeneration, inflammatory response, and neointimal hyperplasia are major pathological processes occurring during in-stent restenosis (ISR). MicroRNAs exhibit great potential in regulating several vascular biological events in different cell types and have been identified as novel therapeutic targets as well as biomarkers for ISR prevention. This review summarized recent experimental and clinical studies on the role of miRNAs in ISR modification, with the aim of unraveling the underlying mechanism and potential therapeutic strategy of ISR.
Collapse
|
6
|
Li H, Song G, Tian W, Ding M, Sun X, Xu J, Dong F, Wang A, Ning P, Yin Y, Wang J. Motility and function of smooth muscle cells in a silk small-caliber tubular scaffold after replacement of rabbit common carotid artery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 114:110977. [DOI: 10.1016/j.msec.2020.110977] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/06/2020] [Accepted: 04/15/2020] [Indexed: 12/17/2022]
|
7
|
Baart VM, Houvast RD, de Geus-Oei LF, Quax PHA, Kuppen PJK, Vahrmeijer AL, Sier CFM. Molecular imaging of the urokinase plasminogen activator receptor: opportunities beyond cancer. EJNMMI Res 2020; 10:87. [PMID: 32725278 PMCID: PMC7387399 DOI: 10.1186/s13550-020-00673-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023] Open
Abstract
The urokinase plasminogen activator receptor (uPAR) plays a multifaceted role in almost any process where migration of cells and tissue-remodeling is involved such as inflammation, but also in diseases as arthritis and cancer. Normally, uPAR is absent in healthy tissues. By its carefully orchestrated interaction with the protease urokinase plasminogen activator and its inhibitor (plasminogen activator inhibitor-1), uPAR localizes a cascade of proteolytic activities, enabling (patho)physiologic cell migration. Moreover, via the interaction with a broad range of cell membrane proteins, like vitronectin and various integrins, uPAR plays a significant, but not yet completely understood, role in differentiation and proliferation of cells, affecting also disease progression. The implications of these processes, either for diagnostics or therapeutics, have received much attention in oncology, but only limited beyond. Nonetheless, the role of uPAR in different diseases provides ample opportunity to exploit new applications for targeting. Especially in the fields of oncology, cardiology, rheumatology, neurology, and infectious diseases, uPAR-targeted molecular imaging could offer insights for new directions in diagnosis, surveillance, or treatment options.
Collapse
Affiliation(s)
- V M Baart
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - R D Houvast
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - L F de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Biomedical Photonic Imaging Group, University of Twente, Enschede, The Netherlands
| | - P H A Quax
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - P J K Kuppen
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - A L Vahrmeijer
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - C F M Sier
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands. .,Percuros BV, Leiden, The Netherlands.
| |
Collapse
|
8
|
Kyyriäinen J, Tapiala J, Lipponen A, Ekolle Ndode-Ekane X, Pitkänen A. Plau/Plaur double-deficiency did not worsen lesion severity or vascular integrity after traumatic brain injury. Neurosci Lett 2020; 729:134935. [PMID: 32360936 DOI: 10.1016/j.neulet.2020.134935] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/06/2020] [Accepted: 03/25/2020] [Indexed: 12/27/2022]
Abstract
Binding of urokinase-type plasminogen activator receptor (uPAR) to its ligand uPA or to its plasma membrane partner, platelet-derived growth factor receptor β (PDGFRβ), promotes neuroprotection, cell proliferation, and angiogenesis. Following injury, single deficiency in uPA or uPAR leads in increased tissue loss and compromised vascular remodeling. We hypothesized that double-deficiency of uPAR (Plaur) and uPA (Plau) would result in increased lesion area and poor vascular integrity after traumatic brain injury (TBI). TBI was induced by lateral fluid-percussion injury in Plau/Plaur double-knockout (dKO) and wild-type (Wt) mice. The cortical lesion area was quantified in unfolded cortical maps prepared from thionin-stained sections at 4 d or 30 d post-TBI. The density of PDGFRβ+ pericytes and blood vessels was calculated from immunostained sections. Blood-brain barrier leakage was analyzed using ImageJ® from IgG-immunostained sections. Genotype had no effect on the total area of the cortical lesion at 4 d or 30 d post-TBI (p > 0.05) or its progression as the overall lesion area was comparable at 4 d and 30 d post-TBI in both genotypes (p > 0.05). Subfield analysis, however, indicated that damage to the visual cortex at 4 d post-TBI in dKO-TBI mice was 53 % of that in Wt-TBI mice (p < 0.05). Both genotypes had a higher density of PDGFRβ-positive pericytes at 4 d than at 30 d post-TBI (p < 0.05), but no genotype effect was detected between these time-points (p > 0.05). TBI-induced increase in the density of PDGFRβ+ blood vessels at the region adjacent to the lesion core was comparable in both genotypes (p > 0.05). Genotype had no effect on TBI-induced IgG leakage into the perilesional cortical parenchyma (p > 0.05). Contrary to our expectations, Plau/Plaur double-deficiency did not aggravate TBI-related structural outcome.
Collapse
Affiliation(s)
- Jenni Kyyriäinen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland
| | - Jesse Tapiala
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland
| | - Anssi Lipponen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland
| | - Xavier Ekolle Ndode-Ekane
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland
| | - Asla Pitkänen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
9
|
Liu S, Yang Y, Jiang S, Tang N, Tian J, Ponnusamy M, Tariq MA, Lian Z, Xin H, Yu T. Understanding the role of non-coding RNA (ncRNA) in stent restenosis. Atherosclerosis 2018; 272:153-161. [PMID: 29609130 DOI: 10.1016/j.atherosclerosis.2018.03.036] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/08/2018] [Accepted: 03/21/2018] [Indexed: 02/02/2023]
Abstract
Coronary heart disease (CHD) is one of the leading disorders with the highest mortality rate. Percutaneous angioplasty and stent implantation are the currently available standard methods for the treatment of obstructive coronary artery disease. However, the stent being an exogenous substance causes several complications by promoting the proliferation of vascular smooth muscle cells, immune responses and neointima formation after implantation, leading to post-stent restenosis (ISR) and late thrombosis. The prevention of these adverse vascular events is important to achieve long-term proper functioning of the heart after stent implantation. Non-coding ribonucleic acids (ncRNAs) are RNA molecules not translated into proteins, theyhave a great potential in regulating endothelial cell and vascular smooth muscle function as well as inflammatory reactions. In this review, we outline the regulatory functions of different classes of ncRNA in cardiovascular disease and propose ncRNAs as new targets for stent restonosis treatment.
Collapse
Affiliation(s)
- Shaoyan Liu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, 266000, People's Republic of China
| | - Yanyan Yang
- Institue for Translational Medicine, Qingdao University, 266021, People's Republic of China
| | - Shaoyan Jiang
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Qingdao University, 266000, People's Republic of China
| | - Ningning Tang
- Institue for Translational Medicine, Qingdao University, 266021, People's Republic of China
| | - Jiawei Tian
- Department of Emergency, The Affiliated Hospital of Qingdao University, 266000, People's Republic of China
| | - Murugavel Ponnusamy
- Institue for Translational Medicine, Qingdao University, 266021, People's Republic of China
| | - Muhammad Akram Tariq
- Department of Biomolecular Engineering, Jack Baskin School of Engineering, University of California, Santa Cruz, CA, United states
| | - Zhexun Lian
- Department of Cardiology, The Affiliated Hospital of Qingdao University, 266000, People's Republic of China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, 266000, People's Republic of China.
| | - Tao Yu
- Institue for Translational Medicine, Qingdao University, 266021, People's Republic of China.
| |
Collapse
|
10
|
Jang MA, Lee SJ, Baek SE, Park SY, Choi YW, Kim CD. α-Iso-Cubebene Inhibits PDGF-Induced Vascular Smooth Muscle Cell Proliferation by Suppressing Osteopontin Expression. PLoS One 2017; 12:e0170699. [PMID: 28114367 PMCID: PMC5256966 DOI: 10.1371/journal.pone.0170699] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 01/09/2017] [Indexed: 12/31/2022] Open
Abstract
α-Iso-cubebene (ICB) is a dibenzocyclooctadiene lignin contained in Schisandra chinensis (SC), a well-known medicinal herb that ameliorates cardiovascular symptoms. Thus, we examined the effect of ICB on vascular smooth muscle cell (VSMC) proliferation, a key feature of diverse vascular diseases. When VSMCs primary cultured from rat thoracic aorta were stimulated with PDGF (1-10 ng/ml), cell proliferation and osteopontin (OPN) expression were concomitantly up-regulated, but these effects were attenuated when cells were treated with MPIIIB10, a neutralizing monoclonal antibody for OPN. In aortic tissues exposed to PDGF, sprouting VSMC numbers increased, which was attenuated in tissues from OPN-deficient mice. Furthermore, VSMC proliferation and OPN expression induced by PDGF were attenuated dose-dependently by ICB (10 or 30 μg/ml). Reporter assays conducted using OPN promoter-luciferase constructs showed that the promoter region 538-234 bp of the transcription start site was responsible for transcriptional activity enhancement by PDGF, which was significantly inhibited by ICB. Putative binding sites for AP-1 and C/EBPβ in the indicated promoter region were suggested by TF Search, and increased binding of AP-1 and C/EBPβ in PDGF-treated VSMCs was demonstrated using a ChIP assay. The increased bindings of AP-1 and C/EBPβ into OPN promoter were attenuated by ICB. Moreover, the PDGF-induced expression of OPN was markedly attenuated in VSMCs transfected with siRNA for AP-1 and C/EBPβ. These results indicate that ICB inhibit VSMC proliferation by inhibiting the AP-1 and C/EBPβ signaling pathways and thus downregulating OPN expression.
Collapse
Affiliation(s)
- Min A. Jang
- Department of Pharmacology, School of Medicine, Pusan National University, Gyeongnam, Republic of Korea
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Gyeongnam, Republic of Korea
| | - Seung Jin Lee
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Seung Eun Baek
- Department of Pharmacology, School of Medicine, Pusan National University, Gyeongnam, Republic of Korea
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Gyeongnam, Republic of Korea
| | - So Youn Park
- Department of Pharmacology, School of Medicine, Pusan National University, Gyeongnam, Republic of Korea
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Gyeongnam, Republic of Korea
| | - Young Whan Choi
- College of Natural Resources & Life Sciences, Pusan National University, Gyeongnam, Republic of Korea
| | - Chi Dae Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Gyeongnam, Republic of Korea
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Gyeongnam, Republic of Korea
| |
Collapse
|
11
|
Osteopontin plays a key role in vascular smooth muscle cell proliferation via EGFR-mediated activation of AP-1 and C/EBPβ pathways. Pharmacol Res 2016; 108:1-8. [DOI: 10.1016/j.phrs.2016.03.041] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/17/2016] [Accepted: 03/27/2016] [Indexed: 12/15/2022]
|
12
|
ZHANG WENBO, SHU CHANG, LI QUANMING, LI MING, LI XIN. Adiponectin affects vascular smooth muscle cell proliferation and apoptosis through modulation of the mitofusin-2-mediated Ras-Raf-Erk1/2 signaling pathway. Mol Med Rep 2015; 12:4703-4707. [DOI: 10.3892/mmr.2015.3899] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 05/06/2015] [Indexed: 11/06/2022] Open
|
13
|
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: 9.3] [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
|
14
|
miR-200c-SUMOylated KLF4 feedback loop acts as a switch in transcriptional programs that control VSMC proliferation. J Mol Cell Cardiol 2015; 82:201-12. [PMID: 25791170 DOI: 10.1016/j.yjmcc.2015.03.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 02/27/2015] [Accepted: 03/10/2015] [Indexed: 02/07/2023]
Abstract
The regulation of vascular smooth muscle cell (VSMC) proliferation is an important issue because it has major implications for the prevention of pathological vascular conditions. Using microRNA array screen, we found the expression levels of 200 unique miRNAs in hyperplasic tissues. Among them, miR-200c expression substantially was down-regulated. The objective of this work was to assess the function of miR-200c and SUMOylated Krϋppel-like transcription factor 4 (KLF4) in the regulation of VSMC proliferation in both cultured cells and animal models of balloon injury. Under basal conditions, we found that miR-200c inhibited the expression of KLF4 and the SUMO-conjugating enzyme Ubc9. Upon PDGF-BB treatment, Ubc9 interacted with and promoted the SUMOylation of KLF4, which allowed the recruitment of transcriptional corepressors (e.g., nuclear receptor corepressor (NCoR) and HDAC2) to the miR-200c promoter. The reduction in miR-200c levels led to increased target gene expression (e.g., Ubc9 and KLF4), which further repressed miR-200c levels and accelerated VSMC proliferation. These results demonstrate that induction of a miR-200c-SUMOylated KLF4 feedback loop is a significant aspect of the PDGF-BB proliferative response in VSMCs and that targeting Ubc9 represents a novel approach for the prevention of restenosis.
Collapse
|
15
|
Abstract
Myocardin (MYOCD) is a potent transcriptional coactivator that functions primarily in cardiac muscle and smooth muscle through direct contacts with serum response factor (SRF) over cis elements known as CArG boxes found near a number of genes encoding for contractile, ion channel, cytoskeletal, and calcium handling proteins. Since its discovery more than 10 years ago, new insights have been obtained regarding the diverse isoforms of MYOCD expressed in cells as well as the regulation of MYOCD expression and activity through transcriptional, post-transcriptional, and post-translational processes. Curiously, there are a number of functions associated with MYOCD that appear to be independent of contractile gene expression and the CArG-SRF nucleoprotein complex. Further, perturbations in MYOCD gene expression are associated with an increasing number of diseases including heart failure, cancer, acute vessel disease, and diabetes. This review summarizes the various biological and pathological processes associated with MYOCD and offers perspectives to several challenges and future directions for further study of this formidable transcriptional coactivator.
Collapse
Affiliation(s)
- Joseph M Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
16
|
Resveratrol inhibits phenotypic switching of neointimal vascular smooth muscle cells after balloon injury through blockade of Notch pathway. J Cardiovasc Pharmacol 2014; 63:233-9. [PMID: 24603118 DOI: 10.1097/fjc.0000000000000040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Phenotypic switching of vascular smooth muscle cells (VSMCs) plays an initial role in neointimal hyperplasia, the main cause of many occlusive vascular diseases. The aim of this study was to measure the effects of resveratrol (RSV) on the phenotypic transformation of VSMCs and to investigate its mechanism of action. METHODS Cultured VSMCs isolated from rat thoracic aorta were prepared with serum starvation for 72 hours followed by RSV treatment (50-200 μmol/L) and 10% serum stimulation. Male Sprague-Dawley rats, subjected to carotid arteries injury from a balloon catheter, were exposed to intraperitoneal injection of RSV (1 mg/kg) or saline and were killed after 7 or 28 days. RESULTS Compared with cells in the serum-induced group, VSMCs in the RSV or N-[N-(3, 5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) treatment group exhibited significant decreases of proliferation and migration. The total and cytoplasmic Notch-1 levels were declined by RSV, accompanied by a significant increase in smooth muscle α-actin and smooth muscle myosin heavy chain protein. The expression of Notch-1, Jagged-1, Hey-1, and Hey-2 mRNA in balloon-injured arteries at 7 days was decreased by RSV treatment. Arteries from RSV-treated rats showed less neointimal hyperplasia, lower collagen content, and a lower rate of cells positive for proliferating cell nuclear antigen 28 days after injury, compared with saline controls. CONCLUSIONS The results indicate that RSV can attenuate phenotypic switching of VSMCs after arterial injury through inhibition of the Notch pathway.
Collapse
|
17
|
Vorpahl M, Schönhofer-Merl S, Michaelis C, Flotho A, Melchior F, Wessely R. The Ran GTPase-activating protein (RanGAP1) is critically involved in smooth muscle cell differentiation, proliferation and migration following vascular injury: implications for neointima formation and restenosis. PLoS One 2014; 9:e101519. [PMID: 24988324 PMCID: PMC4079658 DOI: 10.1371/journal.pone.0101519] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 06/07/2014] [Indexed: 12/12/2022] Open
Abstract
Differentiation and dedifferentiation, accompanied by proliferation play a pivotal role for the phenotypic development of vascular proliferative diseases (VPD), such as restenosis. Increasing evidence points to an essential role of regulated nucleoporin expression in the choice between differentiation and proliferation. However, whether components of the Ran GTPase cycle, which is of pivotal importance for both nucleocytoplasmic transport and for mitotic progression, are subject to similar regulation in VPD is currently unknown. Here, we show that differentiation of human coronary artery smooth muscle cell (CASMC) to a contractile phenotype by stepwise serum depletion leads to significant reduction of RanGAP1 protein levels. The inverse event, dedifferentiation of cells, was assessed in the rat carotid artery balloon injury model, a well-accepted model for neointima formation and restenosis. As revealed by temporospatial analysis of RanGAP1 expression, neointima formation in rat carotid arteries was associated with a significant upregulation of RanGAP1 expression at 3 and 7 days after balloon injury. Of note, neointimal cells located at the luminal surface revealed persistent RanGAP1 expression, as opposed to cells in deeper layers of the neointima where RanGAP1 expression was less or not detectable at all. To gain first evidence for a direct influence of RanGAP1 levels on differentiation, we reduced RanGAP1 in human coronary artery smooth muscle cells by siRNA. Indeed, downregulation of the essential RanGAP1 protein by 50% induced a differentiated, spindle-like smooth muscle cell phenotype, accompanied by an upregulation of the differentiation marker desmin. Reduction of RanGAP1 levels also resulted in a reduction of mitogen induced cellular migration and proliferation as well as a significant upregulation of the cyclin-dependent kinase inhibitor p27KIP1, without evidence for cellular necrosis. These findings suggest that RanGAP1 plays a critical role in smooth muscle cell differentiation, migration and proliferation in vitro and in vivo. Appropriate modulation of RanGAP1 expression may thus be a strategy to modulate VPD development such as restenosis.
Collapse
Affiliation(s)
- Marc Vorpahl
- Department of Cardiology, HELIOS Klinikum Wuppertal, University Witten/Herdecke, Witten, Germany
- * E-mail: (MV); (RW)
| | - Sabine Schönhofer-Merl
- Deutsches Herzzentrum Muenchen and 1. Medizinische Klinik, Klinikum rechts der Isar, Technische Universitaet Muenchen, Muenchen, Germany
| | - Cornelia Michaelis
- Deutsches Herzzentrum Muenchen and 1. Medizinische Klinik, Klinikum rechts der Isar, Technische Universitaet Muenchen, Muenchen, Germany
| | - Annette Flotho
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ - ZMBH Alliance, Heidelberg, Germany
| | - Frauke Melchior
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ - ZMBH Alliance, Heidelberg, Germany
| | - Rainer Wessely
- Deutsches Herzzentrum Muenchen and 1. Medizinische Klinik, Klinikum rechts der Isar, Technische Universitaet Muenchen, Muenchen, Germany
- Zentrum fuer Herz-Gefaess-Lungenmedizin, Cologne, Germany
- * E-mail: (MV); (RW)
| |
Collapse
|
18
|
Narayanaswamy PB, Hodjat M, Haller H, Dumler I, Kiyan Y. Loss of urokinase receptor sensitizes cells to DNA damage and delays DNA repair. PLoS One 2014; 9:e101529. [PMID: 24987841 PMCID: PMC4079571 DOI: 10.1371/journal.pone.0101529] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 06/06/2014] [Indexed: 11/18/2022] Open
Abstract
DNA damage induced by numerous exogenous or endogenous factors may have irreversible consequences on the cell leading to cell cycle arrest, senescence and cell death. The DNA damage response (DDR) is powerful signaling machinery triggered in response to DNA damage, to provide DNA damage recognition, signaling and repair. Most anticancer drugs induce DNA damage, and DNA repair in turn attenuates therapeutic efficiency of those drugs. Approaches delaying DNA repair are often used to increase efficiency of treatment. Recent data show that ubiquitin-proteasome system is essential for signaling and repair of DNA damage. However, mechanisms providing regulation of proteasome intracellular localization, activity, and recruitment to DNA damage sites are elusive. Even less investigated are the roles of extranuclear signaling proteins in these processes. In this study, we report the involvement of the serine protease urokinase-type plasminogen activator receptor (uPAR) in DDR-associated regulation of proteasome. We show that in vascular smooth muscle cells (VSMC) uPAR activates DNA single strand break repair signaling pathway. We provide evidence that uPAR is essential for functional assembly of the 26S proteasome. We further demonstrate that uPAR mediates DNA damage-induced phosphorylation, nuclear import, and recruitment of the regulatory subunit PSMD6 to proteasome. We found that deficiency of uPAR and PSMD6 delays DNA repair and leads to decreased cell survival. These data may offer new therapeutic approaches for diseases such as cancer, cardiovascular and neurodegenerative disorders.
Collapse
MESH Headings
- Active Transport, Cell Nucleus
- Animals
- Cell Line
- Cell Survival
- Cells, Cultured
- DNA Breaks, Single-Stranded
- DNA Repair
- Gene Deletion
- Humans
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Proteasome Endopeptidase Complex/metabolism
- Receptors, Urokinase Plasminogen Activator/genetics
- Receptors, Urokinase Plasminogen Activator/metabolism
Collapse
Affiliation(s)
| | - Mahshid Hodjat
- Nephrology Department, Hannover Medical School, Hannover, Germany
| | - Hermann Haller
- Nephrology Department, Hannover Medical School, Hannover, Germany
| | - Inna Dumler
- Nephrology Department, Hannover Medical School, Hannover, Germany
| | - Yulia Kiyan
- Nephrology Department, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
19
|
Zheng XL. Myocardin and smooth muscle differentiation. Arch Biochem Biophys 2014; 543:48-56. [DOI: 10.1016/j.abb.2013.12.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 12/15/2013] [Accepted: 12/18/2013] [Indexed: 01/08/2023]
|
20
|
Ovsianikov A, Mironov V, Stampfl J, Liska R. Engineering 3D cell-culture matrices: multiphoton processing technologies for biological and tissue engineering applications. Expert Rev Med Devices 2014; 9:613-33. [DOI: 10.1586/erd.12.48] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
21
|
Kalbasi Anaraki P, Patecki M, Larmann J, Tkachuk S, Jurk K, Haller H, Theilmeier G, Dumler I. Urokinase receptor mediates osteogenic differentiation of mesenchymal stem cells and vascular calcification via the complement C5a receptor. Stem Cells Dev 2013; 23:352-62. [PMID: 24192237 DOI: 10.1089/scd.2013.0318] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Vascular calcification is a severe consequence of several pathological processes with a lack of effective therapy. Recent studies suggest that circulating and resident mesenchymal stem cells (MSC) contribute to the osteogenic program of vascular calcification. Molecular mechanisms underlying MSC osteogenic potential and differentiation remain, however, sparsely explored. We investigated a role for the complement receptor C5aR in these processes. We found that expression of C5aR was upregulated upon differentiation of human MSC to osteoblasts. C5aR inhibition by silencing and specific antagonist impaired osteogenic differentiation. We demonstrate that C5aR expression upon MSC differentiation was regulated by the multifunctional urokinase receptor (uPAR). uPAR targeting by siRNA resulted in complete abrogation of C5aR expression and consequently in the inhibition of MSC-osteoblast differentiation. We elucidated the NFκB pathway as the mechanism utilized by the uPAR-C5aR axis. MSC treatment with the NFκB inhibitor completely blocked the differentiation process. Nuclear translocation of the p65 RelA component of the NFκB complex was induced under osteogenic conditions and impaired by the inhibition of uPAR or C5aR. Dual-luciferase reporter assays demonstrated enhanced NFκB signaling upon MSC differentiation, whereas uPAR and C5aR downregulation lead to inhibition of the NFκB activity. We show involvement of the Erk1/2 kinase in this cascade. In vivo studies in a uPAR/LDLR double knockout mouse model of diet-induced atherosclerosis revealed impaired C5aR expression and calcification in aortic sinus plaques in uPAR(-/-)/LDLR(-/-) versus uPAR(+/+)/LDLR(-/-) control animals. These results suggest that uPAR-C5aR axis via the underlying NFκB transcriptional program controls osteogenic differentiation with functional impact on vascular calcification in vivo.
Collapse
|
22
|
Kiyan Y, Tkachuk S, Hilfiker-Kleiner D, Haller H, Fuhrman B, Dumler I. oxLDL induces inflammatory responses in vascular smooth muscle cells via urokinase receptor association with CD36 and TLR4. J Mol Cell Cardiol 2013; 66:72-82. [PMID: 24239845 DOI: 10.1016/j.yjmcc.2013.11.005] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 10/31/2013] [Accepted: 11/05/2013] [Indexed: 10/26/2022]
Abstract
The pathogenesis of atherosclerosis involves an imbalanced lipid metabolism and a deregulated immune response culminating in chronic inflammation of the arterial wall. Recent studies show that endogenous ligands, such as modified plasma lipoproteins, can trigger pattern recognition receptors (PRR) of innate immunity for cellular and humoral reactions. The underlying molecular pathways remain less explored. In this study, we investigated the mechanisms of inflammatory effects of oxidized low-density lipoproteins (oxLDL) on human primary coronary artery smooth muscle cells (VSMC). We show that already low concentration of oxLDL initiated atherogenic signals triggering VSMC transition to proinflammatory phenotype. oxLDL impaired the expression of contractile proteins and myocardin in VSMC and initiated changes in cell functional responses, including expression of proinflammatory molecules. The effects of oxLDL were abolished by downregulation of the multifunctional urokinase receptor (uPAR). In response to oxLDL uPAR associated with CD36 and TLR4, the two main PRR for both pathogen and endogenous ligands. We demonstrate that uPAR association with CD36 and TLR4 mediated oxLDL-induced and NF-κB-dependent G-CSF and GM-CSF expression and changes in VSMC contractile proteins. uPAR-mediated release of G-CSF and GM-CSF by VSMC affected macrophage behavior and production of MCP-1. We provide evidence for functional relevance of our in vitro findings to in vivo human atherosclerotic tissues. Our data imply uPAR as a part of a PRR cluster interfering structurally and functionally with CD36 and TLR4 and responding to endogenous atherogenic ligands. They further point to specific function of each component of this cluster in mediating the ultimate signaling pattern.
Collapse
Affiliation(s)
- Yulia Kiyan
- Nephrology Department, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Sergey Tkachuk
- Nephrology Department, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Denise Hilfiker-Kleiner
- Cardiology Department, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Hermann Haller
- Nephrology Department, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Bianca Fuhrman
- The Lipid Research Laboratory, Technion Faculty of Medicine, and Rambam Medical Center, Haifa, Israel.
| | - Inna Dumler
- Nephrology Department, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
23
|
Wanjare M, Kusuma S, Gerecht S. Perivascular cells in blood vessel regeneration. Biotechnol J 2013; 8:434-47. [PMID: 23554249 DOI: 10.1002/biot.201200199] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 02/19/2013] [Accepted: 03/05/2013] [Indexed: 12/21/2022]
Abstract
Vascular engineering seeks to design and construct functional blood vessels comprising endothelial cells (ECs) and perivascular cells (PCs), with the ultimate goal of clinical translation. While EC behavior has been extensively investigated, PCs play an equally significant role in the development of novel regenerative strategies, providing functionality and stability to vessels. The two major classes of PCs are vascular smooth muscle cells (vSMCs) and pericytes; vSMCs can be further sub-classified as either contractile or synthetic. The inclusion of these cell types is crucial for successful regeneration of blood vessels. Furthermore, understanding distinctions between vSMCs and pericytes will enable improved therapeutics in a tissue-specific manner. Here we focus on the approaches and challenges facing the use of PCs in vascular regeneration, including their characteristics, stem cell sources, and interactions with ECs. Finally, we discuss biochemical and microRNA (miR) regulators of PC behavior and engineering approaches that mimic various cues affecting PC function.
Collapse
Affiliation(s)
- Maureen Wanjare
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | | | | |
Collapse
|
24
|
Kiyan Y, Kurselis K, Kiyan R, Haller H, Chichkov BN, Dumler I. Urokinase receptor counteracts vascular smooth muscle cell functional changes induced by surface topography. Theranostics 2013; 3:516-26. [PMID: 23843899 PMCID: PMC3706695 DOI: 10.7150/thno.4119] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Current treatments for human coronary artery disease necessitate the development of the next generations of vascular bioimplants. Recent reports provide evidence that controlling cell orientation and morphology through topographical patterning might be beneficial for bioimplants and tissue engineering scaffolds. However, a concise understanding of cellular events underlying cell-biomaterial interaction remains missing. In this study, applying methods of laser material processing, we aimed to obtain useful markers to guide in the choice of better vascular biomaterials. Our data show that topographically treated human primary vascular smooth muscle cells (VSMC) have a distinct differentiation profile. In particular, cultivation of VSMC on the microgrooved biocompatible polymer E-shell induces VSMC modulation from synthetic to contractile phenotype and directs formation and maintaining of cell-cell communication and adhesion structures. We show that the urokinase receptor (uPAR) interferes with VSMC behavior on microstructured surfaces and serves as a critical regulator of VSMC functional fate. Our findings suggest that microtopography of the E-shell polymer could be important in determining VSMC phenotype and cytoskeleton organization. They further suggest uPAR as a useful target in the development of predictive models for clinical VSMC phenotyping on functional advanced biomaterials.
Collapse
Affiliation(s)
- Yulia Kiyan
- 1. Nephrology Department, Hannover Medical School, Carl-Neuberg Str. 1, Hannover 30625, Germany
| | - Kestutis Kurselis
- 2. Nanotechnology Department, Laser Zentrum Hannover e.V., Hollerithallee 8, Hannover 30419, Germany
| | - Roman Kiyan
- 2. Nanotechnology Department, Laser Zentrum Hannover e.V., Hollerithallee 8, Hannover 30419, Germany
| | - Hermann Haller
- 1. Nephrology Department, Hannover Medical School, Carl-Neuberg Str. 1, Hannover 30625, Germany
| | - Boris N. Chichkov
- 2. Nanotechnology Department, Laser Zentrum Hannover e.V., Hollerithallee 8, Hannover 30419, Germany
| | - Inna Dumler
- 1. Nephrology Department, Hannover Medical School, Carl-Neuberg Str. 1, Hannover 30625, Germany
| |
Collapse
|
25
|
Hodjat M, Haller H, Dumler I, Kiyan Y. Urokinase receptor mediates doxorubicin-induced vascular smooth muscle cell senescence via proteasomal degradation of TRF2. J Vasc Res 2012; 50:109-23. [PMID: 23172421 DOI: 10.1159/000343000] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 08/23/2012] [Indexed: 11/19/2022] Open
Abstract
The anthracycline doxorubicin is a widely used effective anti-cancer drug. However, its application and dosage are severely limited due to its cardiotoxicity. The exact mechanisms of doxorubicin-induced cardiotoxic side effects remain poorly understood. Even less is known about the impact of doxorubicin treatment on vascular damage. We found that low doses of doxorubicin induced a senescent response in human primary vascular smooth muscle cells (VSMC). We observed that expression of urokinase receptor (uPAR) was upregulated in response to doxorubicin. Furthermore, the level of uPAR expression played a decisive role in developing doxorubicin-induced senescence. uPAR silencing in human VSMC by means of RNA interference as well as uPAR knockout in mouse VSMC resulted in abrogation of doxorubicin-induced cellular senescence. On the contrary, uPAR overexpression promoted VSMC senescence. We further found that proteasomal degradation of telomeric repeat binding factor 2 (TRF2) mediates doxorubicin-induced VSMC senescence. Our results demonstrate that uPAR controls the ubiquitin-proteasome system in VSMC and regulates doxorubicin-induced TRF2 ubiquitination and proteasomal degradation via this mechanism. Therefore, VSMC senescence induced by low doses of doxorubicin may contribute to vascular damage upon doxorubicin treatment. uPAR-mediated TRF2 ubiquitination and proteasomal degradation are further identified as a molecular mechanism underlying this process.
Collapse
Affiliation(s)
- Mahshid Hodjat
- Nephrology Department, Hannover Medical School, Hannover, Germany
| | | | | | | |
Collapse
|
26
|
|
27
|
Ovsianikov A, Chichkov BN. Three-dimensional microfabrication by two-photon polymerization technique. Methods Mol Biol 2012; 868:311-325. [PMID: 22692619 DOI: 10.1007/978-1-61779-764-4_19] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Two-photon polymerization (2PP) technique is a novel CAD/CAM-based technology allowing the fabrication of any computer-designed 3D structure from a photosensitive polymeric material with a lateral resolution down to 100 nm. The fabrication of highly reproducible scaffold structures for tissue engineering by 2PP is very important for systematic studies of cellular processes and better understanding of in vitro tissue formation. Flexibility of this technology and ability to precisely define 3D construct geometry allow the direct addressing of issues associated with vascularization and patient-specific tissue fabrication. In this chapter, we report on our recent advances in the fabrication of biomedical implants and 3D scaffolds for tissue engineering and regenerative medicine by 2PP technique.
Collapse
Affiliation(s)
- Aleksandr Ovsianikov
- Institut für Werkstoffwissenschaft und Werkstofftechnologie, TU Wien, Wien, Austria.
| | | |
Collapse
|