1
|
de Garnica García MG, Mola Solà L, Pérez-Martínez C, Duocastella Codina L, Molina Crisol M, Gómez Castel A, Pérez de Prado A. Comparative evaluation of local and downstream responses in two commercially available paclitaxel-coated balloons in healthy peripheral arteries of a swine model. Cardiovasc Pathol 2025; 74:107688. [PMID: 39179125 DOI: 10.1016/j.carpath.2024.107688] [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] [Received: 07/04/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 08/26/2024] Open
Abstract
OBJECTIVE To investigate the local, downstream, and systemic effects of 2 different paclitaxel-coated balloons. DESIGN Preclinical study in healthy peripheral arteries of a swine model, with randomized allocation of the distribution of the devices: the test paclitaxel-coated balloon (PCB) (LuminorⓇ), a control PCB (IN.PACTⓇ), and a plain angioplasty balloon (OceanusⓇ), considering single (1×) and overlapping (3×) doses with simple blind histologic analysis. METHODS Twenty animals underwent balloon angioplasty at 1× or 3× doses in the external and internal branches of both femoral arteries and were followed-up for 28 days. Postprocedural and follow-up angiography were carried out. Comprehensive necropsy and histology were used to evaluate the local, downstream and systemic effects. RESULTS Angioplasty was successfully carried out in all animals. Significant protocol deviations appeared in 3 arteries (treated with Oceanus®) without clinical relevance. Those samples were excluded from the analysis. All the animals survived the follow-up period without major clinical issues. Local signs of drug toxicity were less marked with Luminor® than IN.PACT® at 1× dose, including endothelial loss (P = .0828), intima/media inflammation (P = .0004), transmural medial smooth muscle cell (SMC) loss (P = .0016), wall thickness loss (P = .0141), presence of fibrin in the vascular wall (P = .0054), and adventitial inflammation (P = .0080). A similar pattern was observed at the 3× dose for endothelial loss (P = .0011), intima/media inflammation (P < .0001), circumferential SMC loss (P = .0004), medial SMC replacement with proteoglycans (P = .0014), fibrin (P = .0034), and collagen content (P = .0205). Downstream vascular histologic changes were mild although more prevalent in the IN.PACT® 3× group (P = .006). No systemic effects of toxicity were detected in any of the samples analyzed. CONCLUSION Luminor® showed better healing pattern (lower inflammation, and endothelial and muscular loss) than IN.PACT® balloon. The effect was evident at single and triple doses. The prevalence of downstream lesions, albeit low, was higher with the triple dose of IN.PACT® compared with Luminor®.
Collapse
Affiliation(s)
- María Gracia de Garnica García
- Department of Animal Health, Section of Pathology, Veterinary School, University of León, León, Spain; Micros Veterinaria S.L., León, Spain
| | | | - Claudia Pérez-Martínez
- Department of Animal Health, Section of Pathology, Veterinary School, University of León, León, Spain.
| | | | | | | | | |
Collapse
|
2
|
Otsu Y, Kuwabara M, Niitsu R, Yamaguchi T, Kodama T. Clinical characteristics and impact of pseudo-lumen blood flow on long-term vessel dilatation in spontaneous isolated dissection of superior mesenteric/celiac artery. Heart Vessels 2024:10.1007/s00380-024-02433-2. [PMID: 38995316 DOI: 10.1007/s00380-024-02433-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 06/20/2024] [Indexed: 07/13/2024]
Abstract
This study aimed to identify the clinical characteristics associated with spontaneous isolated dissection of superior mesenteric artery/celiac artery (SIDSMA/SIDCA). This observational study, conducted at Toranomon Hospital, Japan between 2009 and 2020, analyzed consecutive SIDSMA/SIDCA cases based on radiology data. The study compared clinical characteristics between symptomatic and asymptomatic patients with SIDSMA/SIDCA and investigated factors related to future vessel dilatation. Among 57 cases (44 SIDSMA, 17 SIDCA, and 4 both), the majority were male (87.7%), nearly half having hypertension (43.9%) and smokers (48.9%). Of those, 17 cases (29.8%) were symptomatic; abdominal pain (94.1%), back pain (23.5%), nausea (17.6%) and fever (5.9%). The symptomatic group was younger (52.6 ± 9.4 versus 67.2 ± 7.9 years, P < 0.001), had higher systolic and mean blood pressure (142.6 ± 20.0 versus 129.5 ± 16.5 mmHg, P = 0.017; 96.1 ± 14.6 versus 88.2 ± 17.7 mmHg, P = 0.038), a higher white blood cell count (9975 ± 5032 versus 6268 ± 1991 /µL, P = 0.012), and a higher LDL cholesterol level at diagnosis (129.7 ± 21.7 versus 87.2 ± 25.6 mg/dL, P = 0.002) than the asymptomatic group. The factors associated with future vessel dilatation included the presence of pseudo-lumen flow in the dissection vessel (73.9% versus 41.4%, p = 0.019) and a larger vessel diameter (13.5 ± 2.4 mm versus 11.5 ± 2.1 mm, p = 0.005) at diagnosis after multiple adjustments, pseudo-lumen flow was a predictor of future vessel dilatation (odds ratio, 4.80; 95% confidence interval, 1.11-20.75; p = 0.036). The study revealed that only 30% of SIDSMA/SIDCA cases were symptomatic. Symptomatic cases were generally younger and exhibited higher blood pressure and elevated white blood cell counts. These findings offer valuable insights for the acute diagnosis of SIDSMA/SIDCA.
Collapse
Affiliation(s)
- Yu Otsu
- Department of Cardiology, Toranomon Hospital, 2-2-2, ToranomonTokyo, Minato, 105-8470, Japan
| | - Masanari Kuwabara
- Department of Cardiology, Toranomon Hospital, 2-2-2, ToranomonTokyo, Minato, 105-8470, Japan
| | - Rieko Niitsu
- Department of Cardiology, Toranomon Hospital, 2-2-2, ToranomonTokyo, Minato, 105-8470, Japan
| | - Tetsuo Yamaguchi
- Department of Cardiology, Toranomon Hospital, 2-2-2, ToranomonTokyo, Minato, 105-8470, Japan
| | - Takahide Kodama
- Department of Cardiology, Toranomon Hospital, 2-2-2, ToranomonTokyo, Minato, 105-8470, Japan.
| |
Collapse
|
3
|
Weston WC, Bind MA, Cascio WE, Devlin RB, Diaz-Sanchez D, Ward-Caviness CK. Accelerated aging and altered subclinical response to ozone exposure in young, healthy adults. ENVIRONMENTAL EPIGENETICS 2024; 10:dvae007. [PMID: 38846065 PMCID: PMC11155485 DOI: 10.1093/eep/dvae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 03/11/2024] [Accepted: 05/02/2024] [Indexed: 06/09/2024]
Abstract
Ozone exposure induces a myriad of adverse cardiopulmonary outcomes in humans. Although advanced age and chronic disease are factors that may exacerbate a person's negative response to ozone exposure, there are no molecular biomarkers of susceptibility. Here, we examine whether epigenetic age acceleration (EAA) is associated with responsiveness to short-term ozone exposure. Using data from a crossover-controlled exposure study (n = 17), we examined whether EAA, as measured in lung epithelial cells collected 24 h after clean air exposure, modifies the observed effect of ozone on autonomic function, cardiac electrophysiology, hemostasis, pulmonary function, and inflammation. EAA was assessed in lung epithelial cells extracted from bronchoalveolar lavage fluids, using the pan-tissue aging clock. We used two analytic approaches: (i) median regression to estimate the association between EAA and the estimated risk difference for subclinical responses to ozone and (ii) a block randomization approach to estimate EAA's effect modification of subclinical responses. For both approaches, we calculated Fisher-exact P-values, allowing us to bypass large sample size assumptions. In median regression analyses, accelerated epigenetic age modified associations between ozone and heart rate-corrected QT interval (QTc) ([Formula: see text]= 0.12, P-value = 0.007) and between ozone and C-reactive protein ([Formula: see text] = -0.18, P = 0.069). During block randomization, the directions of association remained consistent for QTc and C-reactive protein; however, the P-values weakened. Block randomization also revealed that responsiveness of plasminogen activator inhibitor-1 (PAI-1) to ozone exposure was modified by accelerated epigenetic aging (PAI-1 difference between accelerated aging-defined block groups = -0.54, P-value = 0.039). In conclusion, EAA is a potential biomarker for individuals with increased susceptibility to ozone exposure even among young, healthy adults.
Collapse
Affiliation(s)
- William C Weston
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Chapel Hill, NC 27514, USA
- US Department of Energy, Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, USA
| | - Marie Abèle Bind
- Biostatistics Center, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Wayne E Cascio
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Chapel Hill, NC 27514, USA
| | - Robert B Devlin
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Chapel Hill, NC 27514, USA
| | - David Diaz-Sanchez
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Chapel Hill, NC 27514, USA
| | - Cavin K Ward-Caviness
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Chapel Hill, NC 27514, USA
| |
Collapse
|
4
|
Gierig M, Gaziano P, Wriggers P, Marino M. Post-angioplasty remodeling of coronary arteries investigated via a chemo-mechano-biological in silico model. J Biomech 2024; 166:112058. [PMID: 38537368 DOI: 10.1016/j.jbiomech.2024.112058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/13/2024]
Abstract
This work presents the application of a chemo-mechano-biological constitutive model of soft tissues for describing tissue inflammatory response to damage in collagen constituents. The material model is implemented into a nonlinear finite element formulation to follow up a coronary standard balloon angioplasty for one year. Numerical results, compared with available in vivo clinical data, show that the model reproduces the temporal dynamics of vessel remodeling associated with subintimal damage. Such dynamics are bimodular, being characterized by an early tissue resorption and lumen enlargement, followed by late tissue growth and vessel constriction. Applicability of the modeling framework in retrospective studies is demonstrated, and future extension towards prospective applications is discussed.
Collapse
Affiliation(s)
- Meike Gierig
- Institute of Continuum Mechanics, Leibniz University of Hannover, An der Universität 1, 30823 Garbsen, Germany
| | - Pierfrancesco Gaziano
- Department of Civil Engineering and Computer Science Engineering, University of Rome Tor Vergata, Via del Politecnico 1, 00133 Rome, Italy
| | - Peter Wriggers
- Institute of Continuum Mechanics, Leibniz University of Hannover, An der Universität 1, 30823 Garbsen, Germany
| | - Michele Marino
- Department of Civil Engineering and Computer Science Engineering, University of Rome Tor Vergata, Via del Politecnico 1, 00133 Rome, Italy.
| |
Collapse
|
5
|
Han G, Wan S, Dandu C, Zhou D, Ding Y, Ji X, Meng R. Intensive mannitol slow infusion post-stenting may attenuate stenting-related early adverse effects in patients with cerebral venous sinus stenosis. CNS Neurosci Ther 2024; 30:e14350. [PMID: 37424178 PMCID: PMC10848041 DOI: 10.1111/cns.14350] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/18/2023] [Accepted: 06/26/2023] [Indexed: 07/11/2023] Open
Abstract
AIMS To analyze intensive slow mannitol poststenting on attenuating stenting-related early adverse effects in cerebral venous sinus stenosis (CVSS). METHODS This real-world study enrolled subacute or chronic CVSS patients from January 2017 through March 2022 and divided them into DSA only and stenting post-DSA groups. The later group was subdivided into control (without extra mannitol use) and intensive slow mannitol subgroup (immediate extra mannitol 250-500 mL, 2 mL/min infusion post-stenting) after signed informed consent. All data were compared. RESULTS A total of 95 eligible patients entered into final analysis, in which 37 cases underwent DSA only and 58 cases underwent stenting post-DSA. Finally, 28 patients were entered into intensive slow mannitol subgroup and 30 in control. Stenting group vs. DSA group, HIT-6 scores and WBC counts were higher in the former (both p < 0.001). Intensive slow mannitol subgroup vs. control on the third day post-stenting, a statistically significant reductions were noticed in the former on WBC counts (6.19 ± 1.86 × 109 /L vs. 9.59 ± 2.05 × 109 /L); HIT-6 scores (degree of headache) (40.00 (38.00-40.00) vs. 49.00 (41.75-55.25)) and brain edema surrounding the stent on CT maps (17.86% vs.96.67%), all p < 0.001. CONCLUSIONS Stenting-related severe headache, inflammatory biomarkers elevation, and brain edema aggravation can be attenuated by intensive slow mannitol infusion.
Collapse
Affiliation(s)
- Guangyu Han
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Advanced Center of StrokeBeijing Institute for Brain DisordersBeijingChina
- National Center for Neurological Disorders, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Shuling Wan
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Advanced Center of StrokeBeijing Institute for Brain DisordersBeijingChina
- National Center for Neurological Disorders, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Chaitu Dandu
- Department of NeurosurgeryWayne State University School of MedicineDetroitMichiganUSA
| | - Da Zhou
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Advanced Center of StrokeBeijing Institute for Brain DisordersBeijingChina
- National Center for Neurological Disorders, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Yuchuan Ding
- Department of NeurosurgeryWayne State University School of MedicineDetroitMichiganUSA
| | - Xunming Ji
- National Center for Neurological Disorders, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Ran Meng
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Advanced Center of StrokeBeijing Institute for Brain DisordersBeijingChina
- National Center for Neurological Disorders, Xuanwu HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
6
|
Pellow C, Jafari Sojahrood A, Zhao X, Kolios MC, Exner AA, Goertz DE. Synchronous Intravital Imaging and Cavitation Monitoring of Antivascular Focused Ultrasound in Tumor Microvasculature Using Monodisperse Low Boiling Point Nanodroplets. ACS NANO 2024; 18:410-427. [PMID: 38147452 PMCID: PMC10786165 DOI: 10.1021/acsnano.3c07711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/28/2023]
Abstract
Focused ultrasound-stimulated microbubbles can induce blood flow shutdown and ischemic necrosis at higher pressures in an approach termed antivascular ultrasound. Combined with conventional therapies of chemotherapy, immunotherapy, and radiation therapy, this approach has demonstrated tumor growth inhibition and profound synergistic antitumor effects. However, the lower cavitation threshold of microbubbles can potentially yield off-target damage that the polydispersity of clinical agent may further exacerbate. Here we investigate the use of a monodisperse nanodroplet formulation for achieving antivascular effects in tumors. We first develop stable low boiling point monodisperse lipid nanodroplets and examine them as an alternative agent to mediate antivascular ultrasound. With synchronous intravital imaging and ultrasound monitoring of focused ultrasound-stimulated nanodroplets in tumor microvasculature, we show that nanodroplets can trigger blood flow shutdown and do so with a sharper pressure threshold and with fewer additional events than conventionally used microbubbles. We further leverage the smaller size and prolonged pharmacokinetic profile of nanodroplets to allow for potential passive accumulation in tumor tissue prior to antivascular ultrasound, which may be a means by which to promote selective tumor targeting. We find that vascular shutdown is accompanied by inertial cavitation and complex-order sub- and ultraharmonic acoustic signatures, presenting an opportunity for effective feedback control of antivascular ultrasound.
Collapse
Affiliation(s)
- Carly Pellow
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
| | - Amin Jafari Sojahrood
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
- Department
of Physics, Toronto Metropolitan University, Toronto M5B 2K3, Canada
- Institute
for Biomedical Engineering, Science and Technology (iBEST), a partnership
between St. Michael’s Hospital, a site of Unity Health Toronto
and Toronto Metropolitan University, Toronto M5B 1T8, Canada
| | - Xiaoxiao Zhao
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
- Department
of Medical Biophysics, University of Toronto, Toronto M5G 1L7, Canada
| | - Michael C. Kolios
- Department
of Physics, Toronto Metropolitan University, Toronto M5B 2K3, Canada
- Institute
for Biomedical Engineering, Science and Technology (iBEST), a partnership
between St. Michael’s Hospital, a site of Unity Health Toronto
and Toronto Metropolitan University, Toronto M5B 1T8, Canada
| | - Agata A. Exner
- Department
of Radiology, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - David E. Goertz
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
- Department
of Medical Biophysics, University of Toronto, Toronto M5G 1L7, Canada
| |
Collapse
|
7
|
Wan H, Li Y, Qin Y, An Y, Yan H, Liu X, Zhang H, Hu C, Li L, Fu D, Yang Y, Dai Y, Luo R, Yang L, Zhang B, Wang Y. Polyphenol-mediated sandwich-like coating promotes endothelialization and vascular healing. Biomaterials 2023; 302:122346. [PMID: 37832504 DOI: 10.1016/j.biomaterials.2023.122346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 09/27/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023]
Abstract
Drug-eluting stents have become one of the most effective methods to treat cardiovascular diseases. However, this therapeutic strategy may lead to thrombosis, stent restenosis, and intimal hyperplasia and prevent re-endothelialization. In this study, we selected 3-aminophenylboronic acid-modified hyaluronic acid and carboxylate chitosan as polyelectrolyte layers and embedded an epigallocatechin-3-gallate-tanshinone IIA sulfonic sodium (EGCG-TSS) complex to develop a sandwich-like layer-by-layer coating. The introduction of a functional molecular EGCG-TSS complex improved not only the biocompatibility of the coating but also its stability by enriching the interaction between the polyelectrolyte coatings through electrostatic interactions, hydrogen bonding, π-π stacking, and covalent bonding. We further elucidated the effectiveness of sandwich-like coatings in regulating the inflammatory response, smooth muscle cell growth behavior, stent thrombosis and restenosis suppression, and vessel re-endothelialization acceleration via in vivo and in vitro. Conclusively, we demonstrated that sandwich-like coating assisted by an EGCG-TSS complex may be an effective surface modification strategy for cardiovascular therapeutic applications.
Collapse
Affiliation(s)
- Huining Wan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yanyan Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yumei Qin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yongqi An
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Hui Yan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Xiyu Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Hao Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Cheng Hu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Linhua Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Daihua Fu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yuan Yang
- Sichuan Xingtai Pule Medical Technology Co Ltd, Chengdu, Sichuan, 610045, China
| | - Yan Dai
- Sichuan Xingtai Pule Medical Technology Co Ltd, Chengdu, Sichuan, 610045, China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Bo Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China.
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| |
Collapse
|
8
|
Hsieh YK, Liu CH, Hu CH. Optimizing Traumatic Limb Salvage: Ectopic Implantation and Staged Rotationplasty. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1879. [PMID: 37893597 PMCID: PMC10608738 DOI: 10.3390/medicina59101879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/10/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023]
Abstract
Rotationplasty, a limb-saving procedure involving a 180-degree ankle rotation to function as a knee joint, is now standard for treating distal femur osteosarcoma. However, challenges related to self-identification persist within the Asian population. This study presents a case involving the successful application of temporary ectopic implantation followed by staged rotationplasty after a severe traumatic amputation, resulting in a favorable outcome. Additionally, a systematic review is conducted to summarize the various difficulties and complications encountered in different studies. This approach improves the feasibility of rotationplasty in traumatic cases and enhances patient and family comprehension.
Collapse
Affiliation(s)
- Yi-Keng Hsieh
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Chang Gung Medical College, Chang Gung University, Taoyuan City 333, Taiwan
| | - Chang-Heng Liu
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City 333, Taiwan
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City 333, Taiwan
| | - Ching-Hsuan Hu
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Chang Gung Medical College, Chang Gung University, Taoyuan City 333, Taiwan
| |
Collapse
|
9
|
Li K, Peng J, Liu Y, Zhang F, Wu D, Luo R, Du Z, Yang L, Liu G, Wang Y. Surface Engineering of Central Venous Catheters via Combination of Antibacterial Endothelium-Mimicking Function and Fibrinolytic Activity for Combating Blood Stream Infection and Thrombosis. Adv Healthc Mater 2023; 12:e2300120. [PMID: 37166220 DOI: 10.1002/adhm.202300120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/09/2023] [Indexed: 05/12/2023]
Abstract
Long-term blood-contacting devices (e.g., central venous catheters, CVCs) still face the highest incidence of blood stream infection and thrombosis in clinical application. To effectively address these complications, this work reports a dual-functional surface engineering strategy for CVCs by organic integration of endothelium-mimicking and fibrinolytic functions. In this proposal, a lysine (Lys)/Cu2+ -incorporated zwitterionic polymer coating (defined as PDA/Lys/Cu-SB) is designed and robustly fabricated onto commercial CVCs using a facile two-step process. Initially, adhesive ene-functionalized dopamine is covalently reacted with Lys and simultaneously coordinated with bactericidal Cu2+ ions, leading to the deposition of a PDA/Lys/Cu coating on CVCs through mussel foot protein inspired surface chemistry. Next, zwitterionic poly(sulfobetaine methacrylate) (pSB) brushes are grafted onto the PDA/Lys/Cu coating to endow lubricant and antifouling properties. In the final PDA/Lys/Cu-SB coating, endothelium-mimicking function is achieved by combining the catalytic generation of nitric oxide from the chelated Cu2+ with antifouling pSB brushes, which led to significant prevention of thrombosis, and bacterial infection in vivo. Furthermore, the immobilized Lys with fibrinolytic activity show remarkably enhanced long-term anti-thrombogenic properties as evidenced in vivo by demonstrating the capability to lyse nascent clots. Therefore, this developed strategy provides a promising solution for long-term blood-contacting devices to combat thrombosis and infection.
Collapse
Affiliation(s)
- Kaijun Li
- College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Jinyu Peng
- College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Yuqi Liu
- College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Fanjun Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Dimeng Wu
- Chengdu Daxan Innovative Medical Tech. Co., Ltd., Chengdu, 611135, China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Zongliang Du
- College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Gongyan Liu
- College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
10
|
Bai Y, Zhang L, Zheng B, Zhang X, Zhang H, Zhao A, Yu J, Yang Z, Wen J. circACTA2 inhibits NLRP3 inflammasome-mediated inflammation via interacting with NF-κB in vascular smooth muscle cells. Cell Mol Life Sci 2023; 80:229. [PMID: 37498354 PMCID: PMC10374705 DOI: 10.1007/s00018-023-04840-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/30/2023] [Accepted: 06/14/2023] [Indexed: 07/28/2023]
Abstract
circACTA2 derived from the smooth muscle α-actin gene plays an important role in the regulation of vascular smooth muscle cell (VSMC) phenotype. The activation of NLRP3 inflammasome is involved in VSMC phenotypic switching. However, the mechanistic relationship between circACTA2 and NLRP3 inflammasome during vascular remodeling remains poorly understood. Here, we showed that circACTA2 was down-regulated in human intimal hyperplasia. circACTA2 overexpression in circACTA2 transgenic mice significantly decreased the neointimal hyperplasia induced by vascular injury, which is concomitant with a decrease in IL-18, IL-1β, TNF-α, and IL-6 levels. Gain- and loss-of-function studies revealed that circACTA2 alleviated VSMC inflammation by suppressing the activation of NLRP3 inflammasome. Mechanistically, circACTA2 inhibited the expression of NF-κB p65 and p50 subunits and interacted with p50, which impedes the formation of the p50/p65 heterodimer and nuclear translocation induced by TNF-α, thus resulting in the suppression of NLRP3 gene transcription and inflammasome activation. Furthermore, circACTA2 overexpression mitigated inflammation via repressing NLRP3 inflammasome-mediated VSMC pyroptosis. Importantly, employing a decoy oligonucleotide to compete with circACTA2 for binding to p50 could attenuate the expression of NLRP3, ASC, and caspase-1. These findings provide a novel insight into the functional roles of circACTA2 in VSMCs, and targeting the circACTA2-NF-κB-NLRP3 axis represents a promising therapeutic strategy for vascular remodeling.
Collapse
Affiliation(s)
- Yang Bai
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017 China
| | - Long Zhang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017 China
| | - Bin Zheng
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017 China
| | - Xinhua Zhang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017 China
- Institution of Chinese Integrative Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017 China
| | - Hong Zhang
- Molecular Biology Laboratory, Talent and Academic Exchange Center, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050017 China
| | - Anning Zhao
- Department of Urology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050017 China
| | - Jing Yu
- Department of Respiratory, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050017 China
| | - Zhan Yang
- Molecular Biology Laboratory, Talent and Academic Exchange Center, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050017 China
| | - Jinkun Wen
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017 China
| |
Collapse
|
11
|
Gierig M, Wriggers P, Marino M. Arterial tissues and their inflammatory response to collagen damage: A continuum in silico model coupling nonlinear mechanics, molecular pathways, and cell behavior. Comput Biol Med 2023; 158:106811. [PMID: 37011434 DOI: 10.1016/j.compbiomed.2023.106811] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/03/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023]
Abstract
Damage in soft biological tissues causes an inflammatory reaction that initiates a chain of events to repair the tissue. This work presents a continuum model and its in silico implementation that describe the cascade of mechanisms leading to tissue healing, coupling mechanical as well as chemo-biological processes. The mechanics is described by means of a Lagrangian nonlinear continuum mechanics framework and follows the homogenized constrained mixtures theory. Plastic-like damage, growth and remodeling as well as homeostasis are taken into account. The chemo-biological pathways account for two molecular and four cellular species, and are activated by damage of collagen molecules in fibers. To consider proliferation, differentiation, diffusion and chemotaxis of species, diffusion-advection-reaction equations are employed. To the best of authors' knowledge, the proposed model combines for the first time such high number of chemo-mechano-biological mechanisms in a consistent continuum biomechanical framework. The resulting set of coupled differential equations describe balance of linear momentum, evolution of kinematic variables as well as mass balance equations. They are discretized in time according to a backward Euler finite difference scheme, and in space through a finite element Galerkin discretization. The features of the model are firstly demonstrated presenting the species dynamics and highlighting the influence of damage intensities on the growth outcome. In terms of a biaxial test, the chemo-mechano-biological coupling and the model's applicability to reproduce normal as well as pathological healing are shown. A last numerical example underlines the model's applicability to complex loading scenarios and inhomogeneous damage distributions. Concluding, the present work contributes towards comprehensive in silico models in biomechanics and mechanobiology.
Collapse
Affiliation(s)
- Meike Gierig
- Institute of Continuum Mechanics, Leibniz University of Hannover, An der Universität 1, 30823 Garbsen, Germany.
| | - Peter Wriggers
- Institute of Continuum Mechanics, Leibniz University of Hannover, An der Universität 1, 30823 Garbsen, Germany
| | - Michele Marino
- Department of Civil Engineering and Computer Science, University of Rome Tor Vergata, Via del Politecnico 1, 00133 Rome, Italy
| |
Collapse
|
12
|
Abstract
Vascular age is determined by functional and structural changes in the arterial wall. When measured by its proxy, pulse wave velocity, it has been shown to predict cardiovascular and total mortality. Disconcordance between chronological and vascular age might represent better or worse vascular health. Cell senescence is caused by oxidative stress and sustained cell replication. Senescent cells acquire senescence-associated secretory phenotype. Oxidative stress, endothelial dysfunction, dysregulation of coagulation and leucocyte infiltration are observed in the aging endothelium. All of these mechanisms lead to increased vascular calcification and stiffness. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can involve the vascular endothelium. It enters cells using angiotensin-converting enzyme 2 (ACE-2) receptors, which are abundant in endothelial cells. The damage this virus does to the endothelium can be direct or indirect. Indirect damage is caused by hyperinflammation. Direct damage results from effects on ACE-2 receptors. The reduction of ACE-2 levels seen during coronavirus disease 2019 (COVID-19) infection might cause vasoconstriction and oxidative stress. COVID-19 and vascular aging share some pathways. Due to the novelty of the virus, there is an urgent need for studies that investigate its long-term effects on vascular health.
Collapse
Affiliation(s)
- Ignas Badaras
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania,Ignas Badaras, Faculty of Medicine, Vilnius
University, M. K. Ciurlionio g. 21/27, LT-03101, Vilnius 01513, Lithuania.
| | - Agnė Laučytė-Cibulskienė
- Department of Nephrology, Skåne University
Hospital, Malmö, Sweden,Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
13
|
Carretta MD, Creutzburg P, Borquez K, Quiroga J, Alarcón P, Rivera A, Burgos RA. Hydroxycarboxylic acid receptor 2 (HCA2) agonists induce NET formation and MMP-9 release from bovine polymorphonuclear leukocytes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 139:104562. [PMID: 36183839 DOI: 10.1016/j.dci.2022.104562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/27/2022] [Accepted: 09/26/2022] [Indexed: 06/16/2023]
Abstract
Periparturient cows are commonly fed diets supplemented with Niacin (nicotinic acid, NA) because of its anti-lipolytic properties. NA confers its anti-lipolytic effects by activating the hydroxycarboxylic acid 2 receptor (HCA2). HCA2 is also activated by the ketone body beta-hydroxybutyrate (BHB) and circulating BHB levels are elevated in postpartum dairy cows. The HCA2 receptor is highly expressed in bovine polymorphonuclear leukocytes (PMN) and could link metabolic and innate immune responses in cattle. We investigated how HCA2 agonists affected bovine PMN function in vitro. We studied different PMN responses, such as granule release, surface expression of CD11b and CD47, generation of neutrophil extracellular traps (NETs), and apoptosis. NA, BHB, and 4,4aR,5,5aR-tetrahydro-1H-cyclopropa [4,5] cyclopenta [1,2-c] pyrazole-3-carboxylic acid (MK-1903) treatment triggered the release of matrix metalloproteinase 9 (MMP-9), a component of the tertiary granule, from neutrophils. Additionally, all HCA2 agonists induced NETs formation but did not affect surface expression of CD11b and CD47. Finally, none of the HCA2 agonists triggered apoptosis in bovine PMN. This information will give new insights into the potential role of the HCA2 receptor in the bovine innate immune response.
Collapse
Affiliation(s)
- Maria Daniella Carretta
- Laboratory of Inflammation Pharmacology, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Science, Universidad Austral de, Chile.
| | - Paz Creutzburg
- Laboratory of Inflammation Pharmacology, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Science, Universidad Austral de, Chile
| | - Katherine Borquez
- Laboratory of Inflammation Pharmacology, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Science, Universidad Austral de, Chile
| | - John Quiroga
- Laboratory of Inflammation Pharmacology, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Science, Universidad Austral de, Chile
| | - Pablo Alarcón
- Laboratory of Inflammation Pharmacology, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Science, Universidad Austral de, Chile
| | - Andrés Rivera
- Laboratory of Inflammation Pharmacology, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Science, Universidad Austral de, Chile
| | - Rafael Agustin Burgos
- Laboratory of Inflammation Pharmacology, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Science, Universidad Austral de, Chile
| |
Collapse
|
14
|
Liu L, Liu P, Yang Y, Dai S, Wang Z, Zhao A, Huang N, Chen J, Yang P. Dual-catalytic CuTPP/TiO 2 nanoparticles for surface catalysis engineering of cardiovascular materials. Mater Today Bio 2022; 17:100494. [PMID: 36425925 PMCID: PMC9678768 DOI: 10.1016/j.mtbio.2022.100494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Endowing materials with catalytic activities analogous to those of the natural endothelium to thus enhance their biological performance has become an option for constructing advanced blood-contact materials. The electron transfer between Cu2+ and Cu+ in the porphyrin center can catalyze the reaction of GSH and GSNO to generate NO, and this electron transfer can also catalyze the decomposition of ROS. Based on this, we created a dual-catalytic surface possessing NO-generating and ROS-scavenging activities to better mimic the versatile catalytic abilities of the endothelium. Copper tetraphenylporphyrin/titanium dioxide nanoparticles (CuTPP/TiO2-NPs) exhibiting excellent NO-generating and ROS-scavenging activities were synthesized and immobilized on the material surface to form a dual-catalytic film (CuTPP/TiO2-film) with the help of the catechol chemistry technique. Unlike most single catalytic surfaces, the dual-catalytic CuTPP/TiO2-film effectively regulated the microenvironment surrounding the implanted device by releasing NO signaling molecules and scavenging harmful ROS. This dual-catalytic film exhibited excellent biosafety and biocompatibility with anti-thrombosis, vascular wall cells (ECs and SMCs) modulation, and anti-inflammatory properties. We envision that this dual-catalytic endothelial bionic strategy may provide a promising solution to the clinical problems plaguing blood-contact devices and provide a novel basis for the further development of surface catalytic-engineered biomaterials.
Collapse
Affiliation(s)
- Luying Liu
- Key Laboratory of Advanced Technology for Materials of the Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Peng Liu
- Key Laboratory of Advanced Technology for Materials of the Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Youhe Yang
- Key Laboratory of Advanced Technology for Materials of the Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Sheng Dai
- Key Laboratory of Advanced Technology for Materials of the Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Zhixing Wang
- Key Laboratory of Advanced Technology for Materials of the Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Ansha Zhao
- Key Laboratory of Advanced Technology for Materials of the Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Nan Huang
- Key Laboratory of Advanced Technology for Materials of the Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Jiang Chen
- Key Laboratory of Advanced Technology for Materials of the Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
- The Department of Ophthalmology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32 of the West Second Section of First Ring Road, Chengdu 610031, PR China
| | - Ping Yang
- Key Laboratory of Advanced Technology for Materials of the Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| |
Collapse
|
15
|
Wang H, Tian Y, Zhang Q, Liu W, Meng L, Jiang X, Xin Y. Essential role of Nrf2 in sulforaphane-induced protection against angiotensin II-induced aortic injury. Life Sci 2022; 306:120780. [PMID: 35839861 DOI: 10.1016/j.lfs.2022.120780] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 12/14/2022]
Abstract
AIMS Cardiovascular disease (CVD) is the leading cause of death worldwide. Inflammation and oxidative stress are the primary factors underlying angiotensin II (Ang II)-induced aortic damage. Nuclear factor erythroid 2-related factor 2 (Nrf2) is an important antioxidative stress factor. Sulforaphane (SFN), which is naturally found in cruciferous vegetables, is an Nrf2 agonist that is safe for oral administration. Here, we aimed to explore the potential of SFN in protecting against Ang II-induced aortic damage by upregulating Nrf2 expression via the extracellular signal-regulated kinase (ERK)/glycogen synthase kinase-3 beta (GSK-3β)/Fyn pathway. MAIN METHODS AND KEY FINDINGS Wild-type (WT) C57BL/6J and Nrf2-knockout (Nrf2-KO) mice were injected with Ang II to induce aortic inflammation, oxidative stress, and cardiac remodeling (increased fibrosis and wall thickness). SFN treatment prevented aortic damage via Nrf2 activation in the WT mice. However, the protective effect of SFN on Ang II-induced aortic damage and upregulation of genes downstream of Nrf2 were not observed in Nrf2-KO mice. SFN induced the upregulation of aortic Nrf2 and inhibited the accumulation of ERK, GSK-3β, and Fyn in the nuclei. SIGNIFICANCE These results revealed that Nrf2 plays a central role in protecting against Ang II-induced aortic injury. Furthermore, SFN prevented Ang II-induced aortic damage by activating Nrf2 through the ERK/GSK-3β/Fyn pathway.
Collapse
Affiliation(s)
- Huanhuan Wang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Yuan Tian
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China; Department of Gynecology, The Second Hospital of Jilin University, Changchun 130041, China.
| | - Qihe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Wenyun Liu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Lingbin Meng
- Department of Hematology and Medical Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA.
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| |
Collapse
|
16
|
Tamang HK, Yang R, Song Z, Hsu S, Peng C, Tung Y, Tzeng B, Chen C. Ca v 3.2 T-type calcium channel regulates mouse platelet activation and arterial thrombosis. J Thromb Haemost 2022; 20:1887-1899. [PMID: 35490411 PMCID: PMC9541131 DOI: 10.1111/jth.15745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Cav 3.2 is a T-type calcium channel that causes low-threshold exocytosis. T-type calcium channel blockers reduce platelet granule exocytosis and aggregation. However, studies of the T-type calcium channel in platelets are lacking. OBJECTIVE To examine the expression and role of Cav 3.2 in platelet function. METHODS Global Cav 3.2-/- and platelet-specific Cav 3.2-/- mice and littermate controls were used for this study. Western blot analysis was used to detect the presence of Cav 3.2 and activation of the calcium-responsive protein extracellular signal-regulated kinase (ERK). Fura-2 dye was used to assess platelet calcium. Flow cytometry and light transmission aggregometry were used to evaluate platelet activation markers and aggregation, respectively. FeCl3 -induced thrombosis and a microfluidic flow device were used to assess in vivo and ex vivo thrombosis, respectively. RESULTS Cav 3.2 was expressed in mouse platelets. As compared with wild-type controls, Cav 3.2-/- mouse platelets showed reduced calcium influx. Similarly, treatment with the T-type calcium channel inhibitor Ni2+ decreased the calcium influx in wild-type platelets. As compared with controls, both Cav 3.2-/- and Ni2+ -treated wild-type platelets showed reduced activation of ERK. ATP release, P-selectin exposure, and αIIb β3 activation were reduced in Cav 3.2-/- and Ni2+ -treated wild-type platelets, as was platelet aggregation. On in vivo and ex vivo thrombosis assay, Cav3.2 deletion caused delayed thrombus formation. However, tail bleeding assay showed intact hemostasis. CONCLUSION These results suggest that Cav 3.2 is required for the optimal activation of platelets.
Collapse
Affiliation(s)
- Hem Kumar Tamang
- Taiwan International Graduate Program in Molecular MedicineNational Yang Ming Chiao Tung University and Academia SinicaTaipeiTaiwan
- Institute of Biomedical SciencesAcademia SinicaTaipeiTaiwan
| | - Ruey‐Bing Yang
- Taiwan International Graduate Program in Molecular MedicineNational Yang Ming Chiao Tung University and Academia SinicaTaipeiTaiwan
- Institute of Biomedical SciencesAcademia SinicaTaipeiTaiwan
| | - Zong‐Han Song
- Institute of Biomedical SciencesAcademia SinicaTaipeiTaiwan
| | - Shao‐Chun Hsu
- Institute of Biomedical SciencesAcademia SinicaTaipeiTaiwan
| | | | - Yi‐Chung Tung
- Research Center for Applied SciencesAcademia SinicaTaipeiTaiwan
| | - Bing‐Hsiean Tzeng
- Division of CardiologyFar Eastern Memorial Hospital and Tri‐Service General HospitalNational Defense Medical CenterTaipeiTaiwan
| | - Chien‐Chang Chen
- Taiwan International Graduate Program in Molecular MedicineNational Yang Ming Chiao Tung University and Academia SinicaTaipeiTaiwan
- Institute of Biomedical SciencesAcademia SinicaTaipeiTaiwan
| |
Collapse
|
17
|
Song Y, Wang K, Loor JJ, Jiang Q, Yang Y, Jiang S, Liu S, He J, Feng X, Du X, Lei L, Gao W, Liu G, Li X. β-Hydroxybutyrate inhibits apoptosis in bovine neutrophils through activating ERK1/2 and AKT signaling pathways. J Dairy Sci 2022; 105:3477-3489. [DOI: 10.3168/jds.2021-21259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/17/2021] [Indexed: 11/19/2022]
|
18
|
Pozzo CFSD, Sielski MS, de Campos Vidal B, Werneck CC, Vicente CP. A collagen I derived matricryptin increases aorta vascular wall remodeling after induced thrombosis in mouse. Thromb Res 2021; 209:59-68. [PMID: 34871983 DOI: 10.1016/j.thromres.2021.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 11/14/2021] [Accepted: 11/18/2021] [Indexed: 11/27/2022]
Abstract
Matricryptins are collagen fragments proteolytically released from the extracellular matrix (ECM) with biological activity that can regulate several processes involved in ECM remodeling. Vessel wall matrix reorganization after lesion is important to the recovery of vascular function. This study aimed to analyze the effect of the peptide p1158/59 (Lindsey, 2015) on thrombosis, neointimal formation, and vascular remodeling of C57BL6 mice abdominal aorta. We used a FeCl3 induced vascular injury mice model and analyzed thrombus size, neointima formation, gelatinase activities in situ, re-endothelization, and collagen fibers organization on the arterial wall using polarization microscopy. As result, we observed that 2 days after injury the treatment with p1158/59 increased thrombus size and gelatinase activity, vascular lesion and it did not recover the endothelium loss induced by the chemical injury. We also observed that the peptide increased neointima growth and collagen birefringence, indicating collagen fibers reorganization. It also promoted increased re-endothelization and decreased activity of gelatinases 14 days after injury. Thus, we conclude that the peptide p1158/59 impaired the initial thrombosis recovery 2 days after injury but was able to induce vascular ECM remodeling after 14 days, improving vessel re-endothelization, collagen fibers deposition, and organization.
Collapse
Affiliation(s)
| | - Micheli Severo Sielski
- Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Benedicto de Campos Vidal
- Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Claudio C Werneck
- Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), São Paulo, Brazil
| | - Cristina Pontes Vicente
- Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| |
Collapse
|
19
|
MED1 Deficiency in Macrophages Accelerates Intimal Hyperplasia via ROS Generation and Inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3010577. [PMID: 34853629 PMCID: PMC8629658 DOI: 10.1155/2021/3010577] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/17/2021] [Accepted: 11/07/2021] [Indexed: 11/17/2022]
Abstract
Mediator complex subunit 1 (MED1) is a component of the mediator complex and functions as a coactivator involved in the regulated transcription of nearly all RNA polymerase II-dependent genes. Previously, we showed that MED1 in macrophages has a protective effect on atherosclerosis; however, the effect of MED1 on intimal hyperplasia and mechanisms regulating proinflammatory cytokine production after macrophage MED1 deletion are still unknown. In this study, we report that MED1 macrophage-specific knockout (MED1 ΔMac) mice showed aggravated neointimal hyperplasia, vascular smooth muscle cells (VSMCs), and macrophage accumulation in injured arteries. Moreover, MED1 ΔMac mice showed increased proinflammatory cytokine production after an injury to the artery. After lipopolysaccharide (LPS) treatment, MED1 ΔMac macrophages showed increased generation of reactive oxygen species (ROS) and reduced expression of peroxisome proliferative activated receptor gamma coactivator-1α (PGC1α) and antioxidant enzymes, including catalase and glutathione reductase. The overexpression of PGC1α attenuated the effects of MED1 deficiency in macrophages. In vitro, conditioned media from MED1 ΔMac macrophages induced more proliferation and migration of VSMCs. To explore the potential mechanisms by which MED1 affects inflammation, macrophages were treated with BAY11-7082 before LPS treatment, and the results showed that MED1 ΔMac macrophages exhibited increased expression of phosphorylated-p65 and phosphorylated signal transducer and activator of transcription 1 (p-STAT1) compared with the control macrophages, suggesting the enhanced activation of NF-κB and STAT1. In summary, these data showed that MED1 deficiency enhanced inflammation and the proliferation and migration of VSMCs in injured vascular tissue, which may result from the activation of NF-κB and STAT1 due to the accumulation of ROS.
Collapse
|
20
|
Graft Preservation Solution DuraGraft ® Alleviates Vascular Dysfunction Following In Vitro Ischemia/Reperfusion Injury in Rats. Pharmaceuticals (Basel) 2021; 14:ph14101028. [PMID: 34681252 PMCID: PMC8538682 DOI: 10.3390/ph14101028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 11/17/2022] Open
Abstract
Vascular ischemia/reperfusion injury (IRI) in patients undergoing coronary artery bypass grafting can result in graft failure and the need for repeat revascularization procedures. DuraGraft® has been shown to protect structure and function in saphenous vein grafts against IRI. We compared the effect of DuraGraft® to saline solution on arterial grafts submitted to IRI. Rat thoracic aortic rings were harvested and immediately mounted in organ bath chambers (control, n = 7 rats) or underwent cold ischemic preservation either in saline (IR, n = 9 rats) or DuraGraft® (IR+Dura, n = 9 rats). Vascular function was measured ex vivo and immunohistochemistry was performed. Impaired maximum vasorelaxation (Rmax) to ACh in the IR-group compared to controls was ameliorated by DuraGraft®, indicating an improvement in endothelial function (Rmax to ACh (%): IR + Dura 73 ± 2 vs. IR 48 ± 3, p < 0.05). Additionally, decreased aortic ring sensitivity to ACh (pD2-value: -log 50% maximum response) seen after IR in the saline group was increased by DuraGraft® (pD2 to ACh: IR+Dura 7.1 ± 0.1 vs. IR 6.3 ± 0.2, p < 0.05). Impaired maximum contractile response to phenylephrine and high potassium chloride concentrations in the IR group compared to controls was significantly improved by DuraGraft®. DuraGraft® alleviates vascular dysfunction following IRI by reducing nitro-oxidative stress and the expression of ICAM-1, without leukocytes engagement.
Collapse
|
21
|
Korkmaz-Icöz S, Kocer C, Sayour AA, Kraft P, Benker MI, Abulizi S, Georgevici AI, Brlecic P, Radovits T, Loganathan S, Karck M, Szabó G. The Sodium-Glucose Cotransporter-2 Inhibitor Canagliflozin Alleviates Endothelial Dysfunction Following In Vitro Vascular Ischemia/Reperfusion Injury in Rats. Int J Mol Sci 2021; 22:ijms22157774. [PMID: 34360539 PMCID: PMC8345991 DOI: 10.3390/ijms22157774] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 07/15/2021] [Indexed: 01/10/2023] Open
Abstract
Vascular ischemia/reperfusion injury (IRI) contributes to graft failure and adverse clinical outcomes following coronary artery bypass grafting. Sodium-glucose-cotransporter (SGLT)-2-inhibitors have been shown to protect against myocardial IRI, irrespective of diabetes. We hypothesized that adding canagliflozin (CANA) (an SGLT-2-inhibitor) to saline protects vascular grafts from IRI. Aortic rings from non-diabetic rats were isolated and immediately mounted in organ bath chambers (control, n = 9–10 rats) or underwent cold ischemic preservation in saline, supplemented either with a DMSO vehicle (IR, n = 8–10 rats) or 50µM CANA (IR + CANA, n = 9–11 rats). Vascular function was measured, the expression of 88 genes using PCR-array was analyzed, and feature selection using machine learning was applied. Impaired maximal vasorelaxation to acetylcholine in the IR-group compared to controls was significantly ameliorated by CANA (IR 31.7 ± 3.2% vs. IR + CANA 51.9 ± 2.5%, p < 0.05). IR altered the expression of 17 genes. Ccl2, Ccl3, Ccl4, CxCr4, Fos, Icam1, Il10, Il1a and Il1b have been found to have the highest interaction. Compared to controls, IR significantly upregulated the mRNA expressions of Il1a and Il6, which were reduced by 1.5- and 1.75-fold with CANA, respectively. CANA significantly prevented the upregulation of Cd40, downregulated NoxO1 gene expression, decreased ICAM-1 and nitrotyrosine, and increased PECAM-1 immunoreactivity. CANA alleviates endothelial dysfunction following IRI.
Collapse
Affiliation(s)
- Sevil Korkmaz-Icöz
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.K.); (A.A.S.); (P.K.); (M.I.B.); (S.A.); (P.B.); (S.L.); (M.K.); (G.S.)
- Correspondence: ; Tel.: +49-6221-566246; Fax: +49-6221-564571
| | - Cenk Kocer
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.K.); (A.A.S.); (P.K.); (M.I.B.); (S.A.); (P.B.); (S.L.); (M.K.); (G.S.)
| | - Alex A. Sayour
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.K.); (A.A.S.); (P.K.); (M.I.B.); (S.A.); (P.B.); (S.L.); (M.K.); (G.S.)
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary;
| | - Patricia Kraft
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.K.); (A.A.S.); (P.K.); (M.I.B.); (S.A.); (P.B.); (S.L.); (M.K.); (G.S.)
| | - Mona I. Benker
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.K.); (A.A.S.); (P.K.); (M.I.B.); (S.A.); (P.B.); (S.L.); (M.K.); (G.S.)
| | - Sophia Abulizi
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.K.); (A.A.S.); (P.K.); (M.I.B.); (S.A.); (P.B.); (S.L.); (M.K.); (G.S.)
| | - Adrian-Iustin Georgevici
- Department of Anesthesiology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany;
| | - Paige Brlecic
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.K.); (A.A.S.); (P.K.); (M.I.B.); (S.A.); (P.B.); (S.L.); (M.K.); (G.S.)
| | - Tamás Radovits
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary;
| | - Sivakkanan Loganathan
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.K.); (A.A.S.); (P.K.); (M.I.B.); (S.A.); (P.B.); (S.L.); (M.K.); (G.S.)
- Department of Cardiac Surgery, University Hospital Halle (Saale), 06120 Halle, Germany
| | - Matthias Karck
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.K.); (A.A.S.); (P.K.); (M.I.B.); (S.A.); (P.B.); (S.L.); (M.K.); (G.S.)
| | - Gábor Szabó
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.K.); (A.A.S.); (P.K.); (M.I.B.); (S.A.); (P.B.); (S.L.); (M.K.); (G.S.)
- Department of Cardiac Surgery, University Hospital Halle (Saale), 06120 Halle, Germany
| |
Collapse
|
22
|
Exosomes from Adipose Mesenchymal Stem Cells Overexpressing Stanniocalcin-1 Promote Reendothelialization After Carotid Endarterium Mechanical Injury. Stem Cell Rev Rep 2021; 18:1041-1053. [PMID: 33982245 DOI: 10.1007/s12015-021-10180-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Stanniocalcin-1 (STC-1) is a secreted glycoprotein that participates in the regulation of inflammation, apoptosis, and necrosis. We investigated the reendothelialization effect of exosomes from adipose stem cells (ADSC) overexpressing STC-1 on injured carotid endarterium. METHODS ADSCs were transfected with lentivirus vectors containing pre-STC-1. PHK-26 as molecular probe was used to track the exosomes engulfed by mice arterial endothelial cells (MAEC). The role of STC-1-ADSC-Exosome (S-ADSC-Exo) in MAECs was verified through scratch test and tube forming. Expressions of STC-1 and NLRP3 inflammasome were detected by western blot and quantitative reverse transcription polymerase chain reaction. Reendothelialization effect was inhibited by the antagonist of siRNA targeting STC-1. Carotid endarterium mechanical injury was induced by insertion with a guidewire into the common carotid artery lumen. Carotid arteries were harvested for histological examination, immunofluorescence staining, and Evan's blue staining. RESULTS Transfection of STC-1 significantly enhanced STC-1 levels in ADSCs, their exosomes, and MAECs. Compared with the control group and the ADSC-Exo group, STC-1 enriched exosomes markedly inhibited the expressions of NLRP3, Caspase-1, and IL-1β in MAECs, exhibited good lateral migration capacity, and promoted angiogenesis. Administration of siRNA targeting STC-1 completely abolished down-regulation of NLRP3, Caspase-1, and IL-1β by STC-1 and inhibited effects of S-ADSC-Exo on lateral migration and angiogenesis. In vivo administration of S-ADSC-Exo had reendothelialization effect on post-injury carotid endarterium as evidenced by thinner arterial wall, low-expressed NLRP3 inflammasome, and more living endothelial cells. CONCLUSIONS The reendothelialization effect of exosomes from ADSCs on post-injury carotid endarterium could be enhanced by genetic modification of the exosomes to contain elevated STC-1, possibly through suppression of NLRP3 inflammasome-mediated inflammation.
Collapse
|
23
|
Li M, Zhu H, Hu X, Gao F, Hu X, Cui Y, Wei X, Xie C, Lv G, Zhao Y, Gao Y. TMEM98, a novel secretory protein, promotes endothelial cell adhesion as well as vascular smooth muscle cell proliferation and migration. Can J Physiol Pharmacol 2021; 99:536-548. [PMID: 32893666 DOI: 10.1139/cjpp-2020-0280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Transmembrane protein 98 (TMEM98) is a novel gene, and its function has not been well investigated. In a prior study, we have shown that siRNA-mediated knockdown of TMEM98 inhibited interleukin-8 (IL-8) promoted endothelial cell (EC) adhesion, as well as vascular smooth muscle cell (VSMC) proliferation and migration in the vascular endothelial and smooth muscle cell dysfunction. Herein, we used gain- and loss-of-function approaches combined with biochemical techniques to further explore the role of TMEM98 in the vascular wall cell. The expression and secretion of TMEM98 was increased in cultured human umbilical vein endothelial cells (HUVECs) and VSMCs treated with IL-8 and platelet-derived growth factor-BB (PDGF-BB). Also, PDGF-BB secretion was increased in TMEM98-treated HUVECs and VSMCs. Thus, it appears that TMEM98 and PDGF-BB form a positive feedback loop in potentiation of EC adhesion, as well as VSMC proliferation and migration. Knockdown of TMEM98 mediated by siRNA inhibited PDGF-BB-promoted EC adhesion by downregulating the expression of ICAM-1 and VCAM-1, as well as impaired the proliferation and migration of VSMCs by suppressing the AKT/GSK3β/cyclin D1 signaling pathway and reducing the expression of β-catenin. Hence, TMEM98 promoted EC adhesion by inducing the expression of ICAM-1/VCAM-1 and triggered VSMC proliferation and migration by activating the ERK and AKT/GSK3β signaling pathways. Taken together, TMEM98 may serve as a potential therapeutic target for the clinical treatment of vascular endothelial and smooth muscle cell dysfunction.
Collapse
Affiliation(s)
- Mei Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Hongmei Zhu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaoyan Hu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Fuhua Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xinxin Hu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ying Cui
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, Dalian Medical University, Dalian, China
| | - Xiaoqing Wei
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, Dalian Medical University, Dalian, China
| | - Ce Xie
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, Dalian Medical University, Dalian, China
| | - Guangxin Lv
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ying Zhao
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, Dalian Medical University, Dalian, China
| | - Ying Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, Dalian Medical University, Dalian, China
| |
Collapse
|
24
|
Zheng M, Guo J, Li Q, Yang J, Han Y, Yang H, Yu M, Zhong L, Lu D, Li L, Sun L. Syntheses and characterization of anti-thrombotic and anti-oxidative Gastrodin-modified polyurethane for vascular tissue engineering. Bioact Mater 2021; 6:404-419. [PMID: 32995669 PMCID: PMC7486448 DOI: 10.1016/j.bioactmat.2020.08.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 12/15/2022] Open
Abstract
Vascular grafts must avoid negative inflammatory responses and thrombogenesis to prohibit fibrotic deposition immediately upon implantation and promote the regeneration of small diameter blood vessels (<6 mm inner diameter). Here, polyurethane (PU) elastomers incorporating anti-coagulative and anti-inflammatory Gastrodin were fabricated. The films had inter-connected pores with porosities equal to or greater than 86% and pore sizes ranging from 250 to 400 μm. Incorporation of Gastrodin into PU films resulted in desirable mechanical properties, hydrophilicity, swelling ratios and degradation rates without collapse. The released Gastrodin maintained bioactivity over 21 days as assessed by its anti-oxidative capability. The Gastrodin/PU had better anti-coagulation response (less observable BSA, fibrinogen and platelet adhesion/activation and suppressed clotting in whole blood). Red blood cell compatibility, measured by hemolysis, was greatly improved with 2Gastrodin/PU compared to other Gastrodin/PU groups. Notably, Gastrodin/PU upregulated anti-oxidant factors Nrf2 and HO-1 expression in H2O2 treated HUVECs, correlated with decreasing pro-inflammatory cytokines TNF-α and IL-1β in RAW 264.7 cells. Upon implantation in a subcutaneous pocket, PU was encapsulated by an obvious fibrous capsule, concurrent with a large amount of inflammatory cell infiltration, while Gastrodin/PU induced a thinner fibrous capsule, especially 2Gastrodin/PU. Further, enhanced adhesion and proliferation of HUVECs seeded onto films in vitro demonstrated that 2Gastrodin/PU could help cell recruitment, as evidenced by rapid host cell infiltration and substantial blood vessel formation in vivo. These results indicate that 2Gastrodin/PU has the potential to facilitate blood vessel regeneration, thus providing new insight into the development of clinically effective vascular grafts.
Collapse
Affiliation(s)
- Meng Zheng
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Jiazhi Guo
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Qing Li
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Jian Yang
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yi Han
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Hongcai Yang
- Department of Neurology, The First Affiliated Hospital, Kunming Medical University, Kunming, 650500, China
| | - Mali Yu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Lianmei Zhong
- Department of Neurology, The First Affiliated Hospital, Kunming Medical University, Kunming, 650500, China
| | - Di Lu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Limei Li
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Lin Sun
- Department of Cardiology, The Second Affiliated Hospital, Kunming Medical University, Kunming, 650032, China
| |
Collapse
|
25
|
Maes L, Vastmans J, Avril S, Famaey N. A Chemomechanobiological Model of the Long-Term Healing Response of Arterial Tissue to a Clamping Injury. Front Bioeng Biotechnol 2021; 8:589889. [PMID: 33575250 PMCID: PMC7870691 DOI: 10.3389/fbioe.2020.589889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/29/2020] [Indexed: 11/22/2022] Open
Abstract
Vascular clamping often causes injury to arterial tissue, leading to a cascade of cellular and extracellular events. A reliable in silico prediction of these processes following vascular injury could help us to increase our understanding thereof, and eventually optimize surgical techniques or drug delivery to minimize the amount of long-term damage. However, the complexity and interdependency of these events make translation into constitutive laws and their numerical implementation particularly challenging. We introduce a finite element simulation of arterial clamping taking into account acute endothelial denudation, damage to extracellular matrix, and smooth muscle cell loss. The model captures how this causes tissue inflammation and deviation from mechanical homeostasis, both triggering vascular remodeling. A number of cellular processes are modeled, aiming at restoring this homeostasis, i.e., smooth muscle cell phenotype switching, proliferation, migration, and the production of extracellular matrix. We calibrated these damage and remodeling laws by comparing our numerical results to in vivo experimental data of clamping and healing experiments. In these same experiments, the functional integrity of the tissue was assessed through myograph tests, which were also reproduced in the present study through a novel model for vasodilator and -constrictor dependent smooth muscle contraction. The simulation results show a good agreement with the in vivo experiments. The computational model was then also used to simulate healing beyond the duration of the experiments in order to exploit the benefits of computational model predictions. These results showed a significant sensitivity to model parameters related to smooth muscle cell phenotypes, highlighting the pressing need to further elucidate the biological processes of smooth muscle cell phenotypic switching in the future.
Collapse
Affiliation(s)
- Lauranne Maes
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Julie Vastmans
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Stéphane Avril
- Mines Saint-Etienne, Université de Lyon, Université Jean Monnet, INSERM, Saint-Étienne, France
| | - Nele Famaey
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
26
|
Zhou F, Hua Y, Ji X, Jia L. A systemic review into carotid plaque features as predictors of restenosis after carotid endarterectomy. J Vasc Surg 2020; 73:2179-2188.e4. [PMID: 33253876 DOI: 10.1016/j.jvs.2020.10.084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/29/2020] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Restenosis after carotid endarterectomy (CEA) limits its long-term efficacy for stroke prevention. Thus, it is of utmost importance to identify the factors that predispose a patient to restenosis after CEA. This systemic review aims to survey the current literature regarding restenosis after CEA and discuss the predictive value of carotid plaque features. METHODS A systemic review of studies on the predictive value of carotid plaque features for restenosis after CEA was conducted according to the PRISMA guidelines. PubMed/MEDLINE and Embase databases were searched up to March 20, 2020. Two authors independently extracted the data and assessed the risk of bias with the Quality in Prognosis Studies tool. Given the heterogeneity in the measurement of prognostic factors, types of CEA, and clinical outcomes, a qualitative synthesis was performed. RESULTS Twenty-one articles with a sample size that ranged from 11 to 1203 were included in this systematic review. Based on the presence of calcification in original carotid plaques, two progression patterns of restenosis were hypothesized: patients with calcified plaques may experience a temporary increase in the intima-media thickness (IMT) followed by a decrease in IMT after CEA, whereas patients with noncalcified plaques may experience a gradual increase in IMT after CEA. Accordingly, patients with a high calcium score may have a high restenosis rate within 6 months after CEA and a low restenosis rate thereafter. Thus, the late restenosis rate in patients with uniformly echogenic plaques was lower than that in patients with uniformly echolucent plaques. Pathologically, a lipid-rich, inflammatory carotid plaque is associated with a decreased risk of restenosis within 1 year after CEA, mainly owing to the relatively mild reactive intimal hyperplasia at the surgical site and active inflammation in the remaining media and adventitia. Molecular predictors for restenosis included a Mannose-binding lectin 2 genotype, preoperative C-reactive protein, serum homocysteine, apolipoprotein J, vitamin C, and telomere length of carotid plaques. CONCLUSIONS This review demonstrated that carotid plaque features, including imaging features, cellular composition, and molecular features, are correlated with the risk of restenosis after CEA. A comprehensive evaluation of plaque characteristics may help to stratify the risk of restenosis after CEA.
Collapse
Affiliation(s)
- Fubo Zhou
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China; Center of Vascular Ultrasonography, Beijing Institute of Brain Disorders, Beijing, China
| | - Yang Hua
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China; Center of Vascular Ultrasonography, Beijing Institute of Brain Disorders, Beijing, China
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Lingyun Jia
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China; Center of Vascular Ultrasonography, Beijing Institute of Brain Disorders, Beijing, China
| |
Collapse
|
27
|
Guo S, Zhang Z, Wang L, Yuan L, Bao J, Zhou J, Jing Z. Six-month results of stenting of the femoropopliteal artery and predictive value of interleukin-6: Comparison with high-sensitivity C-reactive protein. Vascular 2020; 28:715-721. [PMID: 32408853 DOI: 10.1177/1708538120921005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES To determine the association of pre- and postinterventional serum levels of interleukin-6 and high-sensitivity C-reactive protein at the six-month evaluation of restenosis after stenting of the femoropopliteal artery. METHODS Sixty-eight consecutive patients with steno-occlusive femoropopliteal artery disease of Rutherford category III or IV who underwent stent implantation were included. Six-month patency was evaluated with color-coded duplex ultrasound. The association of in-stent restenosis with interleukin-6 and high-sensitivity C-reactive protein levels at baseline, and 24-h postintervention was assessed with a multivariate logistic regression analysis. RESULTS In-stent restenosis was found in 15 patients (22.1%) within six months. Interleukin-6 and high-sensitivity C-reactive protein levels were significantly increased at 24-h postintervention compared to their preintervention values (p < 0.001 and p = 0.002, respectively). Interleukin-6 values at baseline (odds ratio, 1.11; 95% confidence interval: 1.00, 1.23; p = 0.044) and 24-h postintervention (odds ratio, 1.04; 95% confidence interval: 1.02, 1.06; p < 0.001) were independently associated with six-month in-stent restenosis. Twenty-four-hour postinterventional high-sensitivity C-reactive protein levels were also found to be related to restenosis (odds ratio, 1.15; 95% confidence interval: 1.04, 1.26; p = 0.006), but high-sensitivity C-reactive protein levels at baseline did not show an independent association with in-stent restenosis (odds ratio, 0.57; 95% confidence interval: 0.35, 1.80; p = 0.667). Smoking, diabetes mellitus, and cumulative stent length were other parameters associated with an increased risk for in-stent restenosis. CONCLUSIONS Femoropopliteal artery angioplasty with stent placement induces an inflammatory response. Interleukin-6 is a powerful independent predictor of intermediate-term outcomes for stenting of the femoropopliteal artery, suggesting that its predictive value may be superior to that of high-sensitivity C-reactive protein.
Collapse
Affiliation(s)
- Songlin Guo
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhang Zhang
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Lei Wang
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Liangxi Yuan
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Junmin Bao
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jian Zhou
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zaiping Jing
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
28
|
Yang Y, Gao P, Wang J, Tu Q, Bai L, Xiong K, Qiu H, Zhao X, Maitz MF, Wang H, Li X, Zhao Q, Xiao Y, Huang N, Yang Z. Endothelium-Mimicking Multifunctional Coating Modified Cardiovascular Stents via a Stepwise Metal-Catechol-(Amine) Surface Engineering Strategy. RESEARCH (WASHINGTON, D.C.) 2020; 2020:9203906. [PMID: 32405627 PMCID: PMC7196174 DOI: 10.34133/2020/9203906] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/22/2020] [Indexed: 12/14/2022]
Abstract
Stenting is currently the major therapeutic treatment for cardiovascular diseases. However, the nonbiogenic metal stents are inclined to trigger a cascade of cellular and molecular events including inflammatory response, thrombogenic reactions, smooth muscle cell hyperproliferation accompanied by the delayed arterial healing, and poor reendothelialization, thus leading to restenosis along with late stent thrombosis. To address prevalence critical problems, we present an endothelium-mimicking coating capable of rapid regeneration of a competently functioning new endothelial layer on stents through a stepwise metal (copper)-catechol-(amine) (MCA) surface chemistry strategy, leading to combinatorial endothelium-like functions with glutathione peroxidase-like catalytic activity and surface heparinization. Apart from the stable nitric oxide (NO) generating rate at the physiological level (2.2 × 10-10 mol/cm2/min lasting for 60 days), this proposed strategy could also generate abundant amine groups for allowing a high heparin conjugation efficacy up to ∼1 μg/cm2, which is considerably higher than most of the conventional heparinized surfaces. The resultant coating could create an ideal microenvironment for bringing in enhanced anti-thrombogenicity, anti-inflammation, anti-proliferation of smooth muscle cells, re-endothelialization by regulating relevant gene expressions, hence preventing restenosis in vivo. We envision that the stepwise MCA coating strategy would facilitate the surface endothelium-mimicking engineering of vascular stents and be therefore helpful in the clinic to reduce complications associated with stenosis.
Collapse
Affiliation(s)
- Ying Yang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane 4059, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane 4059, Australia
| | - Peng Gao
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Juan Wang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Qiufen Tu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Long Bai
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane 4059, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane 4059, Australia
| | - Kaiqin Xiong
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Hua Qiu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xin Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Manfred F. Maitz
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
- Max Bergmann Center of Biomaterials, Leibniz Institute of Polymer Research Dresden, Hohe Strasse 6, 01069 Dresden, Germany
| | - Huaiyu Wang
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiangyang Li
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Yin Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane 4059, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane 4059, Australia
| | - Nan Huang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Zhilu Yang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
29
|
Mottola G, Werlin EC, Wu B, Chen M, Chatterjee A, Schaller MS, Conte MS. Oral Resolvin D1 attenuates early inflammation but not intimal hyperplasia in a rat carotid angioplasty model. Prostaglandins Other Lipid Mediat 2019; 146:106401. [PMID: 31841663 DOI: 10.1016/j.prostaglandins.2019.106401] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 08/19/2019] [Accepted: 11/26/2019] [Indexed: 12/16/2022]
Abstract
Inflammation ensuing from vascular injury promotes intimal hyperplasia (IH) and restenosis. Resolvin D1 (RvD1) is a lipid mediator that attenuates IH in vivo when delivered locally to the vessel wall in animal models. We tested the hypothesis that peri-procedural oral administration of RvD1 could blunt the local inflammatory response to angioplasty, and attenuate downstream IH. Carotid angioplasty was performed on rats fed with either RvD1 or vehicle through oral gavage, starting one day prior to injury until post-operative day (POD) 3 or 14 when arteries were harvested. To study pharmacokinetics and bioactivity of oral RvD1, we measured plasma RvD1 by ELISA, whole blood phagocytosis activity using flow cytometry, and cAMP levels in the thoracic aorta by ELISA. Carotid arteries were harvested on POD3 for staining (anti-CD45, anti-Myeloperoxidase (MPO), anti-Ki67 or dihydroethidium (DHE) for reactive oxygen species), mRNA expression of target genes (quantitative RT-PCR), or on POD14 for morphometry (elastin stain). RvD1 plasma concentration peaked 3 h after gavage in rats, at which point we concurrently observed an increase in circulating monocyte phagocytosis activity and aortic cAMP levels in RvD1-treated rats vs. vehicle. Oral RvD1 attenuated local arterial inflammation after angioplasty by reducing CD45+, MPO+, Ki67+ cells, and DHE staining intensity. Oral RvD1 also reduced the expression of several pro-inflammatory genes within the injured vessels. However, oral RvD1 did not significantly reduce IH. Oral RvD1 attenuated acute inflammation within the arterial wall after angioplasty in rats, but did not significantly affect IH.
Collapse
Affiliation(s)
- Giorgio Mottola
- Department of Surgery, Division of Vascular and Endovascular Surgery, University of California San Francisco, Cardiovascular Research Institute, 555 Mission Bay Blvd South, San Francisco, 94143, CA, USA
| | - Evan C Werlin
- Department of Surgery, Division of Vascular and Endovascular Surgery, University of California San Francisco, Cardiovascular Research Institute, 555 Mission Bay Blvd South, San Francisco, 94143, CA, USA
| | - Bian Wu
- Department of Surgery, Division of Vascular and Endovascular Surgery, University of California San Francisco, Cardiovascular Research Institute, 555 Mission Bay Blvd South, San Francisco, 94143, CA, USA
| | - Mian Chen
- Department of Surgery, Division of Vascular and Endovascular Surgery, University of California San Francisco, Cardiovascular Research Institute, 555 Mission Bay Blvd South, San Francisco, 94143, CA, USA
| | - Anuran Chatterjee
- Department of Surgery, Division of Vascular and Endovascular Surgery, University of California San Francisco, Cardiovascular Research Institute, 555 Mission Bay Blvd South, San Francisco, 94143, CA, USA
| | - Melinda S Schaller
- Department of Surgery, Division of Vascular and Endovascular Surgery, University of California San Francisco, Cardiovascular Research Institute, 555 Mission Bay Blvd South, San Francisco, 94143, CA, USA
| | - Michael S Conte
- Department of Surgery, Division of Vascular and Endovascular Surgery, University of California San Francisco, Cardiovascular Research Institute, 555 Mission Bay Blvd South, San Francisco, 94143, CA, USA.
| |
Collapse
|
30
|
Buckley MR, Terry PD, Kirkpatrick SS, Arnold JD, McNally MM, Grandas OH, Freeman MB, Goldman MH, Whelan J, Mountain DJ. Dietary supplementation with Zyflamend poly-herbal extracts and fish oil inhibits intimal hyperplasia development following vascular intervention. Nutr Res 2019; 68:34-44. [PMID: 31306903 DOI: 10.1016/j.nutres.2019.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 10/26/2022]
Abstract
The polyherbal blend Zyflamend™ has been shown to have anti-inflammatory properties and attenuate inflammatory-modulated pathologies. Fish oils have also been shown to have cardioprotective properties. However, the beneficial effects of their combination have not been investigated. Intimal hyperplasia (IH), a pathological remodeling response of a vessel to injury, is heavily regulated by an immune-mediated reaction. The objective of this study was to determine if dietary supplementation with Zyflamend and/or Wholemega could affect inflammatory-dependent vascular remodeling mechanisms when provided at human equivalent doses. Based on their anti-inflammatory properties and protective benefits demonstrated in previous pre-clinical studies, we hypothesized administration of these supplements would prevent IH in an animal model of vascular injury. The diets of aged male rats were supplemented with human equivalent doses of Zyflamend (Zyf) and/or Wholemega (WMega) or placebo (Plac) for 1wk prior to balloon angioplasty (BA)-induced injury of the left carotid artery. At 28d post-injury morphometric analysis of carotid tissue revealed IH was decreased in Zyf + WMega animals compared to placebo, while Zyf or WMega independently had no significant effect. Serum cytokine screening indicated injury-induced interleukin family isoforms, interferon-γ, and macrophage inflammatory proteins were downregulated by Zyf + WMega. Immunohistochemical staining for monocyte/macrophage phenotypic markers revealed that while overall monocyte/macrophage vessel infiltration was not affected, Zyf + WMega limited the alternative differentiation of M2 macrophages and reduced the presence of myofibroblasts in the injured vessel wall. In summary, dietary supplementation with Zyf + WMega attenuated the acute inflammatory response following vascular injury and inhibited IH development in vivo.
Collapse
Affiliation(s)
- Michael R Buckley
- University of Tennessee Graduate School of Medicine, Department of Surgery, Knoxville, TN
| | - Paul D Terry
- University of Tennessee Graduate School of Medicine, Department of Medicine, Knoxville, TN
| | - Stacy S Kirkpatrick
- University of Tennessee Graduate School of Medicine, Department of Surgery, Knoxville, TN
| | - Joshua D Arnold
- University of Tennessee Graduate School of Medicine, Department of Surgery, Knoxville, TN
| | - Michael M McNally
- University of Tennessee Graduate School of Medicine, Department of Surgery, Knoxville, TN
| | - Oscar H Grandas
- University of Tennessee Graduate School of Medicine, Department of Surgery, Knoxville, TN
| | - Michael B Freeman
- University of Tennessee Graduate School of Medicine, Department of Surgery, Knoxville, TN
| | - Mitchell H Goldman
- University of Tennessee Graduate School of Medicine, Department of Surgery, Knoxville, TN
| | - Jay Whelan
- University of Tennessee Knoxville, Department of Nutrition, Knoxville, TN
| | - Deidra Jh Mountain
- University of Tennessee Graduate School of Medicine, Department of Surgery, Knoxville, TN.
| |
Collapse
|
31
|
Ammann KR, DeCook KJ, Li M, Slepian MJ. Migration versus proliferation as contributor to in vitro wound healing of vascular endothelial and smooth muscle cells. Exp Cell Res 2019; 376:58-66. [PMID: 30660619 PMCID: PMC6988716 DOI: 10.1016/j.yexcr.2019.01.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/31/2018] [Accepted: 01/10/2019] [Indexed: 12/27/2022]
Abstract
Wound closure, as a result of collective cell growth, is an essential biological response to injury. In the field of vascular biology, the response of vascular smooth muscle cells (SMCs) and endothelial cells (ECs) to injury and substrate surface is important in therapeutic clinical treatment interventions such as angioplasty and atherectomy. Specifically, the mechanism by which cells close wounds (i.e. proliferation versus migration) in response to injury stimuli is of interest to better modulate recurrent vascular stenosis, prevent thrombus formation, occlusion, and life-threatening cardiovascular events. Here, we examine growth extent and temporal sequence of events following wound or gap introduction to a confluent monolayer of vascular SMCs or ECs. Significant differences in the preferred mechanisms of these cells to close wounds or gaps were observed; after 48 h, 73% of SMC wound closure was observed to be due to proliferation, while 75% of EC wound closure resulted from migration. These mechanisms were further modulated via addition or removal of extracellular matrix substrate and injury, with ECs more responsive to substrate composition and less to injury, in comparison to SMCs. Our results indicate that ECs and SMCs heal wounds differently, and that the time and mode of injury and associated substrate surface all impact this response.
Collapse
Affiliation(s)
- Kaitlyn R Ammann
- Department of Biomedical Engineering, College of Engineering, University of Arizona,1127 E James E Rogers Way, PO Box 210020, Tucson, AZ 85721, USA
| | - Katrina J DeCook
- Department of Biomedical Engineering, College of Engineering, University of Arizona,1127 E James E Rogers Way, PO Box 210020, Tucson, AZ 85721, USA
| | - Maxwell Li
- Department of Biomedical Engineering, College of Engineering, University of Arizona,1127 E James E Rogers Way, PO Box 210020, Tucson, AZ 85721, USA
| | - Marvin J Slepian
- Department of Biomedical Engineering, College of Engineering, University of Arizona,1127 E James E Rogers Way, PO Box 210020, Tucson, AZ 85721, USA; Department of Medicine, Sarver Heart Center, University of Arizona, 1501 N Campbell Ave, PO Box 245035, Tucson, AZ 85724, USA.
| |
Collapse
|
32
|
Yi B, Shen Y, Tang H, Wang X, Li B, Zhang Y. Stiffness of Aligned Fibers Regulates the Phenotypic Expression of Vascular Smooth Muscle Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:6867-6880. [PMID: 30676736 DOI: 10.1021/acsami.9b00293] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Electrospun uniaxially aligned ultrafine fibers show great promise in constructing vascular grafts mimicking the anisotropic architecture of native blood vessels. However, understanding how the stiffness of aligned fibers would impose influences on the functionality of vascular cells has yet to be explored. The present study aimed to explore the stiffness effects of electrospun aligned fibrous substrates (AFSs) on phenotypic modulation in vascular smooth muscle cells (SMCs). A stable jet coaxial electrospinning (SJCES) method was employed to generate highly aligned ultrafine fibers of poly(l-lactide- co-caprolactone)/poly(l-lactic acid) (PLCL/PLLA) in shell-core configuration with a remarkably varying stiffness region from 0.09 to 13.18 N/mm. We found that increasing AFS stiffness had no significant influence on the cellular shape and orientation along the fiber direction with the cultured human umbilical artery SMCs (huaSMCs) but inhibited the cell adhesion rate, promoted cell proliferation and migration, and especially enhanced the F-actin fiber assembly in the huaSMCs. Notably, higher fiber stiffness resulted in significant downregulation of contractile markers like alpha-smooth muscle actin (α-SMA), smooth muscle myosin heavy chain, calponin, and desmin, whereas upregulated the gene expression of pathosis-associated osteopontin ( OPN) in the huaSMCs. These results allude to the phenotype of huaSMCs on stiffer AFSs being miserably modulated into a proliferative and pathological state. Consequently, it adversely affected the proliferation and migration behavior of human umbilical vein endothelial cells as well. Moreover, stiffer AFSs also revealed to incur significant upregulation of inflammatory gene expression, such as interleukin-6 ( IL-6), monocyte chemoattractant protein-1 ( MCP-1), and intercellular adhesion molecule-1 ( ICAM-1), in the huaSMCs. This study stresses that although electrospun aligned fibers are capable of modulating native-like oriented cell morphology and even desired phenotype realization or transition, they might not always direct cells into correct functionality. The integrated fiber stiffness underlying is thereby a critical parameter to consider in engineering structurally anisotropic tissue-engineered vascular grafts to ultimately achieve long-term patency.
Collapse
Affiliation(s)
| | | | | | | | - Bin Li
- Department of Orthopaedics , The First Affiliated Hospital of Soochow University , Suzhou 215006 , China
- Orthopaedic Institute, Medical College , Soochow University , Suzhou 215007 , China
- China Orthopaedic Regenerative Medicine Group (CORMed) , Hangzhou 310058 , China
| | - Yanzhong Zhang
- China Orthopaedic Regenerative Medicine Group (CORMed) , Hangzhou 310058 , China
| |
Collapse
|
33
|
Mo X, Yan F, Zhang B. Molecular Ultrasound Monitoring of Early Artery Injury After Carotid Balloon Angioplasty. Front Pharmacol 2019; 9:1569. [PMID: 30740054 PMCID: PMC6355681 DOI: 10.3389/fphar.2018.01569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 12/24/2018] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular intervention is a common treatment procedure for many cardiovascular diseases. But restenosis often occurs after these procedures, greatly discounting their long-term therapeutic effects. Early detection of endothelial denudation is helpful for the diagnosis and prevention of restenosis. Here, we fabricated targeted microbubbles by conjugating anti-collagen IV antibodies to the surface of biotinylated microbubbles (MBColIV) and applied them for ultrasound molecular imaging of endothelial injury at early stage. Our results showed that the MBColIV, with a typical multi-peak particle distribution, was successfully constructed, which was confirmed by Alexa Fluor® 555-labeled secondary antibody. Ex vivo adhesion of microbubbles revealed that MBColIV can effectively and specially bind to the surface of balloon-injured carotid artery. The in vivo animal experiments showed ultrasound molecular imaging signals from carotid artery-injured rats administrated with MBColIV were significantly higher than those administrated with isotype control microbubbles. Histological staining of the left carotid common artery revealed that collagen IV was obviously exposed after endothelium denudation in balloon-injured artery. In conclusion, our current study provides an effective approach to detect vascular injury at the early stage and a potential platform for image-guided therapy to vascular injury.
Collapse
Affiliation(s)
- Xinhai Mo
- Department of Ultrasound in Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fei Yan
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bo Zhang
- Department of Ultrasound in Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
Grad E, Zolotarevsky K, Danenberg HD, Nordling-David MM, Gutman D, Golomb G. The role of monocyte subpopulations in vascular injury following partial and transient depletion. Drug Deliv Transl Res 2018; 8:945-953. [PMID: 28656488 DOI: 10.1007/s13346-017-0404-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The innate immunity system plays a critical role in vascular repair and restenosis development. Liposomes encapsulating bisphosphonates (LipBPs), but not free BPs, suppress neointima formation following vascular injury mediated in part by monocytes. The objective of this study was to elucidate the role of monocyte subpopulations on vascular healing following LipBP treatment. The potency- and dose-dependent treatment effect of clodronate (CLOD) and alendronate (ALN) liposomes on restenosis inhibition, total monocyte depletion, and monocytes subpopulation was studied. Rats subjected to carotid injury were treated by a single IV injection of LipBPs at the time of injury. Low- and high-dose LipALN treatment (3 and 10 mg/kg, respectively) resulted in a dose-dependent effect on restenosis development after 30 days. Both doses of LipALN resulted in a dose-dependent inhibition of restenosis, but only high dose of LipALN depleted monocytes (-60.1 ± 4.4%, 48 h post injury). Although LipCLOD treatment (at an equivalent potency to 3 mg/kg alendronate) significantly reduced monocyte levels (72.1 ± 6%), no restenosis inhibition was observed. The major finding of this study is the correlation found between monocyte subclasses and restenosis inhibition. Non-classical monocyte (NCM) levels were found higher in LipALN-treated rats, but lower in LipCLOD-treated rats, 24 h after injury and treatment. We suggest that the inhibition of circulating monocyte subpopulations is the predominant mechanism by which LipBPs prevent restenosis. The effect of LipBP treatment on the monocyte subpopulation correlates with the dose and potency of LipBPs.
Collapse
Affiliation(s)
- Etty Grad
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 12065 Ein Kerem Medical Cenre, 91120, Jerusalem, Israel
| | - Ksenia Zolotarevsky
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 12065 Ein Kerem Medical Cenre, 91120, Jerusalem, Israel
| | - Haim D Danenberg
- Cardiovascular Research Center, Hadassah Hebrew University Medical Center, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Mirjam M Nordling-David
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 12065 Ein Kerem Medical Cenre, 91120, Jerusalem, Israel
| | - Dikla Gutman
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 12065 Ein Kerem Medical Cenre, 91120, Jerusalem, Israel
| | - Gershon Golomb
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 12065 Ein Kerem Medical Cenre, 91120, Jerusalem, Israel.
| |
Collapse
|
35
|
Mediating the invasion of smooth muscle cells into a cell-responsive hydrogel under the existence of immune cells. Biomaterials 2018; 180:193-205. [DOI: 10.1016/j.biomaterials.2018.07.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 05/27/2018] [Accepted: 07/11/2018] [Indexed: 01/12/2023]
|
36
|
Genetic lineage tracing analysis of c-kit + stem/progenitor cells revealed a contribution to vascular injury-induced neointimal lesions. J Mol Cell Cardiol 2018; 121:277-286. [PMID: 30053526 DOI: 10.1016/j.yjmcc.2018.07.252] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 07/23/2018] [Indexed: 11/23/2022]
Abstract
AIMS Accumulating evidence indicates the presence of vascular stem/progenitor cells that may play a role in endothelial repair and lesion formation in the injured artery, in which c-kit+ stem/progenitor cells have been reported to differentiate into endothelial and smooth muscle cells in vitro and in ischemic tissue. In this study, we investigated whether and how endogenous c-kit+ stem/progenitor cells contribute to vascular injury and neointima formation in vivo. METHODS AND RESULTS We created Kit-CreERxRosa26-RFP mice and performed genetic lineage tracing analysis of c-kit+ stem/progenitor cells in injury-induced neointima formation in vivo. We provide direct evidence that endogenous c-kit+ stem/progenitor cells minimally differentiate into endothelial or smooth muscle cells facilitating vascular repair, but predominantly generate monocytes/macrophages and granulocytes contributing to vascular immuno-inflammatory response to endothelial injury. Although c-kit+ cells reside in both bone marrow and vessel wall, bone marrow transplantation data indicate that bone marrow-derived c-kit+ cells are the main source for enhancing neointima formation. Furthermore, treatment of ACK2, a c-kit receptor antagonizer, attenuates neointimal hyperplasia after injury at least in part by depleting c-kit+ cells and their generated progeny. CONCLUSIONS c-kit+ stem/progenitor cells are not a main source for endothelial regeneration and smooth muscle accumulation of the large artery injury, but a plausible interventional approach to reduce vascular immuno-inflammatory response and subsequently to ameliorate vascular lesions.
Collapse
|
37
|
Liu G, Gong Y, Zhang R, Piao L, Li X, Liu Q, Yan S, Shen Y, Guo S, Zhu M, Yin H, Funk CD, Zhang J, Yu Y. Resolvin E1 attenuates inj ury‐induced vascular neointimal formation by inhibition of inflammatory responses and vascular smooth muscle cell migration. FASEB J 2018; 32:5413-5425. [DOI: 10.1096/fj.201800173r] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Guizhu Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
- Department of Pharmacology, College of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Yanjun Gong
- CAS Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
| | - Rui Zhang
- Department of Pharmacology, College of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Lingjuan Piao
- Graduate School of Pharmaceutical SciencesCollege of Pharmacy, Ewha Women's UniversitySeoulSouth Korea
| | - Xinzhi Li
- Department of Biomedical and Molecular SciencesQueen's UniversityKingston OntarioCanada
| | - Qian Liu
- Department of Pharmacology, College of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Shuai Yan
- CAS Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
| | - Yujun Shen
- Department of Pharmacology, College of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Shumin Guo
- CAS Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
| | - Mingjiang Zhu
- CAS Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
| | - Huiyong Yin
- CAS Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
| | - Colin D. Funk
- Department of Biomedical and Molecular SciencesQueen's UniversityKingston OntarioCanada
| | - Jian Zhang
- Department of Pharmacology, College of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Ying Yu
- CAS Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
- Department of Pharmacology, College of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| |
Collapse
|
38
|
Cuomo F, Coppola A, Botti C, Maione C, Forte A, Scisciola L, Liguori G, Caiafa I, Ursini MV, Galderisi U, Cipollaro M, Altucci L, Cobellis G. Pro-inflammatory cytokines activate hypoxia-inducible factor 3α via epigenetic changes in mesenchymal stromal/stem cells. Sci Rep 2018; 8:5842. [PMID: 29643458 PMCID: PMC5895792 DOI: 10.1038/s41598-018-24221-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 03/23/2018] [Indexed: 11/09/2022] Open
Abstract
Human mesenchymal stromal/stem cells (hMSCs) emerged as a promising therapeutic tool for ischemic disorders, due to their ability to regenerate damaged tissues, promote angiogenesis and reduce inflammation, leading to encouraging, but still limited results. The outcomes in clinical trials exploring hMSC therapy are influenced by low cell retention and survival in affected tissues, partially influenced by lesion's microenvironment, where low oxygen conditions (i.e. hypoxia) and inflammation coexist. Hypoxia and inflammation are pathophysiological stresses, sharing common activators, such as hypoxia-inducible factors (HIFs) and NF-κB. HIF1α and HIF2α respond essentially to hypoxia, activating pathways involved in tissue repair. Little is known about the regulation of HIF3α. Here we investigated the role of HIF3α in vitro and in vivo. Human MSCs expressed HIF3α, differentially regulated by pro-inflammatory cytokines in an oxygen-independent manner, a novel and still uncharacterized mechanism, where NF-κB is critical for its expression. We investigated if epigenetic modifications are involved in HIF3α expression by methylation-specific PCR and histone modifications. Robust hypermethylation of histone H3 was observed across HIF3A locus driven by pro-inflammatory cytokines. Experiments in a murine model of arteriotomy highlighted the activation of Hif3α expression in infiltrated inflammatory cells, suggesting a new role for Hif3α in inflammation in vivo.
Collapse
Affiliation(s)
- Francesca Cuomo
- Department of Biochemistry, Biophysics and General Pathology, Università degli Studi della Campania L. Vanvitelli, Via L. De Crecchio, 7, 80138, Naples, Italy
| | - Antonietta Coppola
- Department of Biochemistry, Biophysics and General Pathology, Università degli Studi della Campania L. Vanvitelli, Via L. De Crecchio, 7, 80138, Naples, Italy
| | - Chiara Botti
- Department of Biochemistry, Biophysics and General Pathology, Università degli Studi della Campania L. Vanvitelli, Via L. De Crecchio, 7, 80138, Naples, Italy
- Laboratorio di Patologia Clinica, Ospedale Santobono, Via M. Fiore 6, 80129, Naples, Italy
| | - Ciro Maione
- Department of Biochemistry, Biophysics and General Pathology, Università degli Studi della Campania L. Vanvitelli, Via L. De Crecchio, 7, 80138, Naples, Italy
| | - Amalia Forte
- Department of Experimental Medicine, Università degli Studi della Campania L. Vanvitelli, Via L. De Crecchio, 7, 80138, Naples, Italy
| | - Lucia Scisciola
- Department of Biochemistry, Biophysics and General Pathology, Università degli Studi della Campania L. Vanvitelli, Via L. De Crecchio, 7, 80138, Naples, Italy
| | - Giuseppina Liguori
- Istituto Nazionale Tumori, Struttura Complessa Oncologia Medica Melanoma Immunoterapia Oncologica e Terapia Innovativa, Via M. Semmola, 80131, Naples, Italy
| | - Ilaria Caiafa
- Department of Biochemistry, Biophysics and General Pathology, Università degli Studi della Campania L. Vanvitelli, Via L. De Crecchio, 7, 80138, Naples, Italy
| | - Matilde Valeria Ursini
- Institute of Genetics and Biophysics, 'A. Buzzati-Traverso' (IGB), via P. Castellino, 111, 80131, Naples, Italy
| | - Umberto Galderisi
- Department of Experimental Medicine, Università degli Studi della Campania L. Vanvitelli, Via L. De Crecchio, 7, 80138, Naples, Italy
| | - Marilena Cipollaro
- Department of Experimental Medicine, Università degli Studi della Campania L. Vanvitelli, Via L. De Crecchio, 7, 80138, Naples, Italy
| | - Lucia Altucci
- Department of Biochemistry, Biophysics and General Pathology, Università degli Studi della Campania L. Vanvitelli, Via L. De Crecchio, 7, 80138, Naples, Italy
| | - Gilda Cobellis
- Department of Biochemistry, Biophysics and General Pathology, Università degli Studi della Campania L. Vanvitelli, Via L. De Crecchio, 7, 80138, Naples, Italy.
| |
Collapse
|
39
|
Kim EJ, Park SY, Baek SE, Jang MA, Lee WS, Bae SS, Kim K, Kim CD. HMGB1 Increases IL-1β Production in Vascular Smooth Muscle Cells via NLRP3 Inflammasome. Front Physiol 2018; 9:313. [PMID: 29643819 PMCID: PMC5882820 DOI: 10.3389/fphys.2018.00313] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 03/14/2018] [Indexed: 12/18/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the major cell type in the blood vessel walls, and their phenotypic modulation is a key cellular event driving vascular remodeling. Although high mobility group box-1 (HMGB1) plays a pivotal role in inflammatory processes after vascular injuries, the importance of the links between VSMCs, HMGB1 and vascular inflammation has not been clarified. To prove the hypothesis that VSMCs might be active players in vascular inflammation by secreting inflammatory cytokines, we investigated the proinflammatory effects of HMGB1 and its intermediary signaling pathways in VSMCs. When cultured human VSMCs were stimulated with HMGB1 (10–500 ng/ml), IL-1β production was markedly increased. HMGB1 also increased the expression of NLRP3 inflammasome components including NLRP3, ASC and caspase-1. Among these components, HMGB1-induced expressions of NLRP3 and caspase-1 were markedly attenuated in TLR2 siRNA-transfected cells, whereas ASC and caspase-1 expressions were reduced in RAGE-deficient cells. In TLR4-deficient cells, HMGB1-induced caspase-1 expression was significantly attenuated. Moreover, IL-1β production in HMGB1-stimulated cells was significantly reduced in cells transfected with caspase-1 siRNA as well as in cells treated with monoclonal antibodies or siRNAs for TLR2, TLR4 and RAGE. Overall, this study identified a pivotal role for NLRP3 inflammasome and its receptor signaling involved in the production of IL-1β in VSMCs stimulated with HMGB1. Thus, targeting HMGB1 signaling in VSMCs offers a promising therapeutic strategy for treating vascular remodeling diseases.
Collapse
Affiliation(s)
- Eun Jung Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, South Korea.,Gene and Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, South Korea
| | - So Youn Park
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, South Korea.,Gene and Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, South Korea
| | - Seung Eun Baek
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, South Korea.,Gene and Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, South Korea
| | - Min A Jang
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, South Korea.,Gene and Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, South Korea
| | - Won Suk Lee
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, South Korea
| | - Sun Sik Bae
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, South Korea.,Gene and Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, South Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, South Korea
| | - Chi Dae Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, South Korea.,Gene and Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, South Korea
| |
Collapse
|
40
|
A Cell-Adhesive Plasma Polymerized Allylamine Coating Reduces the In Vivo Inflammatory Response Induced by Ti6Al4V Modified with Plasma Immersion Ion Implantation of Copper. J Funct Biomater 2017; 8:jfb8030030. [PMID: 28726761 PMCID: PMC5618281 DOI: 10.3390/jfb8030030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/13/2017] [Accepted: 07/17/2017] [Indexed: 01/16/2023] Open
Abstract
Copper (Cu) could be suitable to create anti-infective implants based on Titanium (Ti), for example by incorporating Cu into the implant surface using plasma immersion ion implantation (Cu-PIII). The cytotoxicity of Cu might be circumvented by an additional cell-adhesive plasma polymerized allylamine film (PPAAm). Thus, this study aimed to examine in vivo local inflammatory reactions for Ti6Al4V implants treated with Cu-PIII (Ti-Cu), alone or with an additional PPAAm film (Ti-Cu-PPAAm), compared to untreated implants (Ti). Successful Cu-PIII and PPAAm treatment was confirmed with X-ray Photoelectron Spectroscopy. Storage of Ti-Cu and Ti-Cu-PPAAm samples in double-distilled water for five days revealed a reduction of Cu release by PPAAm. Subsequently, Ti, Ti-Cu and Ti-Cu-PPAAm samples were simultaneously implanted into the neck musculature of 24 rats. After 7, 14 and 56 days, peri-implant tissue was retrieved from 8 rats/day for morphometric immunohistochemistry of different inflammatory cells. On day 56, Ti-Cu induced significantly stronger reactions compared to Ti (tissue macrophages, antigen-presenting cells, T lymphocytes) and to Ti-Cu-PPAAm (tissue macrophages, T lymphocytes, mast cells). The response for Ti-Cu-PPAAm was comparable with Ti. In conclusion, PPAAm reduced the inflammatory reactions caused by Cu-PIII. Combining both plasma processes could be useful to create antibacterial and tissue compatible Ti-based implants.
Collapse
|
41
|
Liu S, Yuan Y, Zhou Y, Zhao M, Chen Y, Cheng J, Lu Y, Liu J. Phloretin attenuates hyperuricemia-induced endothelial dysfunction through co-inhibiting inflammation and GLUT9-mediated uric acid uptake. J Cell Mol Med 2017; 21:2553-2562. [PMID: 28402018 PMCID: PMC5618667 DOI: 10.1111/jcmm.13176] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 02/20/2017] [Indexed: 02/05/2023] Open
Abstract
Hyperuricemia is an important risk factor for cardiovascular and renal diseases. Phloretin had shown antioxidant and anti‐inflammatory properties, but its role in endothelial injury is rarely reported. In this study, we aimed to investigate the protective effect of phloretin on UA‐induced injury in human umbilical vein endothelial cells. The effects of UA and phloretin on cell viability, inflammation, THP‐1 monocyte adhesion, endothelial cell tube formation, GLUT9 expression and UA uptake in human umbilical vein endothelial cells were evaluated. The changes of nuclear factor‐kappa B/extracellular regulated protein kinases signalling were also analysed. Our results showed that UA reduced cell viability and tube formation, and increased inflammation and monocytes adhesion in human umbilical vein endothelial cells in a dose‐dependent manner. In contrast, phloretin significantly attenuated pro‐inflammatory factors expression and endothelial injury induced by UA. Phloretin inhibited the activation of extracellular regulated protein kinases/nuclear factor‐kappa B pathway, and reduced GLUT9 and it mediated UA uptake in human umbilical vein endothelial cells. These results indicated that phloretin attenuated UA‐induced endothelial injury via a synergic mechanism including direct anti‐inflammatory effect and lowering cellular UA uptake. Our study suggested that phloretin might be a promising therapy for hyperuricemia‐related cardiovascular diseases.
Collapse
Affiliation(s)
- Shuyun Liu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yujia Yuan
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yijie Zhou
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Meng Zhao
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Younan Chen
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jingqiu Cheng
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jingping Liu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
42
|
Elshaer SL, Lorys RE, El-Remessy AB. Cell Therapy and Critical Limb Ischemia: Evidence and Window of Opportunity in Obesity. ACTA ACUST UNITED AC 2016; 3. [PMID: 28979948 DOI: 10.15226/2374-8354/3/1/00121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Sally L Elshaer
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia 30912, USA
| | - Renee E Lorys
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia 30912, USA
| | - A B El-Remessy
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia 30912, USA
| |
Collapse
|
43
|
Bledsoe SL, Barr JC, Fitzgerald RT, Brown AT, Faas FH, Eidt JF, Moursi MM. Pravastatin and Clopidogrel Combined Inhibit Intimal Hyperplasia in a Rat Carotid Endarterectomy Model. Vasc Endovascular Surg 2016; 40:49-57. [PMID: 16456606 DOI: 10.1177/153857440604000107] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Intimal hyperplasia, resulting from a complex cascade of events involving platelets, leukocytes, and smooth muscle cells, may be inhibited by the HMG-CoA reductase inhibitor pravastatin, which demonstrates inhibition of platelet activity and leukocyte adhesion and may be associated with inhibition of vascular smooth muscle cell proliferation and migration. Clopidogrel, an adenosine diphosphate (ADP) receptor inhibitor, was shown to decrease platelet activity and aggregation but not intimal hyperplasia (IH). We postulated that the combination of both pravastatin and clopidogrel would significantly decrease IH in a rat carotid endarterectomy model. Male Sprague-Dawley rats (n = 18) divided by treatment regimen underwent treatment for 2 weeks both before and after an open carotid endarterectomy. Serum collected at the time of harvest was measured for C-reactive protein (CRP), platelet activity, and total serum cholesterol; carotid arteries were removed and processed for IH determination. Control rats (n = 7) received oral vehicle daily before and following endarterectomy. Pravastatin-alone rats (n = 6) received oral pravastatin (10 mg/kg/day) before and after endarterectomy. Pravastatin plus clopidogrel rats (n = 5) received oral pravastatin (10 mg/kg/day) plus a preendarterectomy bolus of oral clopidogrel (4.3 mg/kg) before endarterectomy and resumed pravastatin (10 mg/kg/day) plus oral clopidogrel (1 mg/kg/day) postendarterectomy. Pravastatin alone and pravastatin plus clopidogrel significantly decreased CRP compared to controls (120.2 ±11.2 and 134.1 ±9.9 vs 191.1 ±9.2 µg/mL, respectively p = 0.003 and p = 0.0024). CRP levels were not different between pravastatin alone and pravastatin plus clopidogrel (p = 0.35). Platelet activity was significantly decreased by pravastatin alone and pravastatin plus clopidogrel in comparison to controls (7.3 ±2.2 and 6.6 ±2.8 vs 19.2 ±6.1 platelet reactive units (PRU), respectively p = 0.048 and p = 0.045). No significant difference was noted in platelet activity between pravastatin alone and pravastatin plus clopidogrel (p = 0.89). Pravastatin plus clopidogrel significantly reduced serum cholesterol compared to control and pravastatin alone (84.0 ±6.6 vs 110.4 ±7.4 and 117.0 ±8.8 mg/dL, respectively p = 0.03 and p = 0.01). Pravastatin alone did not decrease serum cholesterol compared to controls (p = 0.54). IH was not reduced by pravastatin alone compared to controls (p = 0.61) but was significantly decreased by pravastatin plus clopidogrel in comparison to control and pravastatin alone (3.0 ±1.1 vs 46.3 ±13.7 and 37.4 ±14.6% luminal stenosis, respectively p = 0.01 and p = 0.05). Pravastatin plus clopidogrel significantly decreased CRP, platelet activity, total serum cholesterol, and IH while pravastatin alone decreased only CRP and platelet activity. Intimal hyperplasia reduction may therefore be dependent on other contributors, possibly growth factors, cytokines, and oxidative stress. The combination of pravastatin plus clopidogrel may have synergistic or even additional inhibitory effects on IH. Pravastatin plus clopidogrel was effective in decreasing IH in a rat carotid endarterectomy model and may prove a useful therapy for IH reduction in the clinical setting.
Collapse
Affiliation(s)
- Shelly L Bledsoe
- Department of Surgery, Division of Vascular Surgery, University of Arkansas for Medical Sciences, Central Arkansas Veterans Healthcare System, Little Rock, 72205, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Kukida M, Mogi M, Ohshima K, Nakaoka H, Iwanami J, Kanno H, Tsukuda K, Chisaka T, Min LJ, Wang XL, Bai HY, Shan BS, Higaki A, Yamauchi T, Okura T, Higaki J, Horiuchi M. Angiotensin II Type 2 Receptor Inhibits Vascular Intimal Proliferation With Activation of PPARγ. Am J Hypertens 2016; 29:727-36. [PMID: 26471325 DOI: 10.1093/ajh/hpv168] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/24/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Angiotensin II type 2 (AT2) receptor stimulation could exert beneficial effects on vascular remodeling. Previously, we reported that AT2 receptor stimulation ameliorated insulin resistance in diabetic mice accompanied by PPARγ activation which also plays a variety of crucial roles in the vasculature. Therefore, this study aimed to investigate the vascular protective effect of the AT2 receptor with activation of PPARγ involving AT2 receptor-interacting protein (ATIP). METHODS AND RESULTS Vascular injury was induced by polyethylene-cuff placement around the femoral artery in C57BL/6J mice. Treatment with compound 21 (C21), an AT2 receptor agonist, decreased neointimal formation, cell proliferation, and the mRNA levels of monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor (TNF)-α, and interleukin-1β, and phosphorylation of nuclear factor-kappa B, and increased PPARγ DNA-binding activity in the injured artery, whereas these inhibitory effects of C21 were attenuated by co-treatment with a PPARγ antagonist, GW9662. Treatment of vascular smooth muscle cells (VSMC) with C21 prepared from smAT2 transgenic mice, which highly express the AT2 receptor in VSMC, increased both PPARγ activity and its DNA-binding activity determined by dual-luciferase assay and electrophoresis mobility shift assay (EMSA), respectively. We observed that ATIP was involved in PPARγ complex formation, and that transfection of siRNA of ATIP1 attenuated the AT2 receptor-mediated increase in PPARγ activity in VSMC. In response to AT2 receptor stimulation, ATIP was translocated from the plasma membrane to the nucleus. CONCLUSIONS Our results suggest a new mechanism by which AT2 receptor stimulation activates PPARγ, thereby resulting in amelioration of vascular intimal proliferation, and that ATIP plays an important role in AT2 receptor-mediated PPARγ activation.
Collapse
Affiliation(s)
- Masayoshi Kukida
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan; Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Masaki Mogi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan;
| | - Kousei Ohshima
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan; Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Hirotomo Nakaoka
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Jun Iwanami
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Harumi Kanno
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Kana Tsukuda
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Toshiyuki Chisaka
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan; Department of Pediatrics, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Li-Juan Min
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Xiao-Li Wang
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Hui-Yu Bai
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Bao-Shuai Shan
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Akinori Higaki
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan; Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Toshifumi Yamauchi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan; Department of Pediatrics, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Takafumi Okura
- Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Jitsuo Higaki
- Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Masatsugu Horiuchi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| |
Collapse
|
45
|
Moulakakis KG, Sfyroeras GS, Papapetrou A, Antonopoulos CN, Mantas G, Kakisis J, Alepaki M, Mylonas SN, Karakitsos P, Liapis CD. Inflammatory response and renal function following endovascular repair of the descending thoracic aorta. J Endovasc Ther 2016; 22:201-6. [PMID: 25809362 DOI: 10.1177/1526602815573227] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE To evaluate inflammatory response and renal function after thoracic endovascular aortic repair (TEVAR) of lesions in the descending thoracic aorta. METHODS Thirty-two consecutive patients treated with TEVAR from January 2010 to August 2013 were enrolled in this prospective study. Two were excluded owing to dissecting thoracic aortic aneurysm (TAA) extending into the renal arteries with renal failure in one and a saccular TAA in which a multilayer flow-modulating stent was implanted in the other. This left 30 patients (28 men; mean age 68.8±5.9 years) with 28 TAAs, an aortic dissection, and an aortic ulcer for the analysis. Temperature and serum levels of white blood cells (WBCs), C-reactive protein (CRP), interleukin-10 (IL-10), IL-6, IL-8, tumor necrosis factor-alpha (TNF-α), creatinine, urea, and cystatin C were measured preoperatively and at 24 and 48 hours postoperatively. RESULTS Statistically significant increases in temperature and serum levels of WBCs, CRP, IL-10, and IL-6 were observed 24 and 48 hours postoperatively compared to baseline (all p<0.05). The number of endografts and the coverage of the celiac or subclavian artery did not affect the magnitude of the inflammatory response. No significant differences were observed concerning serum levels of IL-8, TNF-α, creatinine, or cystatin C from baseline to 24 or 48 hours postoperatively. CONCLUSION Endograft implantation in the thoracic aorta may propagate an inflammatory response during the early postoperative period. No clinical adverse events related to the increased inflammatory response were observed. Renal function does not seem to be deteriorated after TEVAR in the descending thoracic aorta.
Collapse
Affiliation(s)
| | | | | | | | - George Mantas
- Attikon University Hospital, University of Athens, Greece
| | - John Kakisis
- Attikon University Hospital, University of Athens, Greece
| | - Maria Alepaki
- Attikon University Hospital, University of Athens, Greece
| | | | | | | |
Collapse
|
46
|
Postolow F, Fediuk J, Nolette N, Hinton M, Dakshinamurti S. Thromboxane promotes smooth muscle phenotype commitment but not remodeling of hypoxic neonatal pulmonary artery. FIBROGENESIS & TISSUE REPAIR 2015; 8:20. [PMID: 26583045 PMCID: PMC4650498 DOI: 10.1186/s13069-015-0037-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 10/20/2015] [Indexed: 12/19/2022]
Abstract
Background Persistent pulmonary hypertension of the newborn (PPHN) is characterized by vasoconstriction and pulmonary vascular remodeling. Remodeling is believed to be a response to physical or chemical stimuli including pro-mitotic inflammatory mediators such as thromboxane. Our objective was to examine the effects of hypoxia and thromboxane signaling ex vivo and in vitro on phenotype commitment, cell cycle entry, and proliferation of PPHN and control neonatal pulmonary artery (PA) myocytes in tissue culture. Methods To examine concurrent effects of hypoxia and thromboxane on myocyte growth, serum-fed first-passage newborn porcine PA myocytes were randomized into normoxic (21 % O2) or hypoxic (10 % O2) culture for 3 days, with daily addition of thromboxane mimetic U46619 (10−9 to 10−5 M) or diluent. Cell survival was detected by MTT assay. To determine the effect of chronic thromboxane exposure (versus whole serum) on activation of arterial remodeling, PPHN was induced in newborn piglets by a 3-day hypoxic exposure (FiO2 0.10); controls were 3 day-old normoxic and day 0 piglets. Third-generation PA were segmented and cultured for 3 days in physiologic buffer, Ham’s F-12 media (in the presence or absence of 10 % fetal calf serum), or media with 10−6 M U46619. DNA synthesis was measured by 3H-thymidine uptake, protein synthesis by 3H-leucine uptake, and proliferation by immunostaining for Ki67. Cell cycle entry was studied by laser scanning cytometry of nuclei in arterial tunica media after propidium iodide staining. Phenotype commitment was determined by immunostaining tunica media for myosin heavy chain and desmin, quantified by laser scanning cytometry. Results Contractile and synthetic myocyte subpopulations had differing responses to thromboxane challenge. U46619 decreased proliferation of synthetic and contractile myocytes. PPHN arteries exhibited decreased protein synthesis under all culture conditions. Serum-supplemented PA treated with U46619 had decreased G1/G0 phase myocytes and an increase in S and G2/M. When serum-deprived, PPHN PA incubated with U46619 showed arrested cell cycle entry (increased G0/G1, decreased S and G2/M) and increased abundance of contractile phenotype markers. Conclusions We conclude that thromboxane does not initiate phenotypic dedifferentiation and proliferative activation in PPHN PA. Exposure to thromboxane triggers cell cycle exit and myocyte commitment to contractile phenotype.
Collapse
Affiliation(s)
- Fabiana Postolow
- Department of Pediatrics, University of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada
| | - Jena Fediuk
- Department of Physiology, University of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada ; Biology of Breathing Group, Manitoba Institute of Child Health, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada
| | - Nora Nolette
- Biology of Breathing Group, Manitoba Institute of Child Health, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada
| | - Martha Hinton
- Biology of Breathing Group, Manitoba Institute of Child Health, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada
| | - Shyamala Dakshinamurti
- Department of Pediatrics, University of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada ; Department of Physiology, University of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada ; Biology of Breathing Group, Manitoba Institute of Child Health, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada ; Section of Neonatology, WS012 Women's Hospital, 735 Notre Dame Ave, Winnipeg, MB R3E 0L8 Canada
| |
Collapse
|
47
|
Moulakakis KG, Mylonas SN, Kakisis J, Geroulakos G. Inflammatory response following stent grafting for acute aortic syndrome. Eur J Cardiothorac Surg 2015; 49:1247-8. [DOI: 10.1093/ejcts/ezv341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
48
|
Rutten B, Roest M, McClellan EA, Sels JW, Stubbs A, Jukema JW, Doevendans PA, Waltenberger J, van Zonneveld AJ, Pasterkamp G, De Groot PG, Hoefer IE. Platelet density per monocyte predicts adverse events in patients after percutaneous coronary intervention. Thromb Haemost 2015; 115:353-60. [PMID: 26423019 DOI: 10.1160/th15-03-0227] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 08/24/2015] [Indexed: 12/24/2022]
Abstract
Monocyte recruitment to damaged endothelium is enhanced by platelet binding to monocytes and contributes to vascular repair. Therefore, we studied whether the number of platelets per monocyte affects the recurrence of adverse events in patients after percutaneous coronary intervention (PCI). Platelet-monocytes complexes with high and low median fluorescence intensities (MFI) of the platelet marker CD42b were isolated using cell sorting. Microscopic analysis revealed that a high platelet marker MFI on monocytes corresponded with a high platelet density per monocyte while a low platelet marker MFI corresponded with a low platelet density per monocyte (3.4 ± 0.7 vs 1.4 ± 0.1 platelets per monocyte, P=0.01). Using real-time video microscopy, we observed increased recruitment of high platelet density monocytes to endothelial cells as compared with low platelet density monocytes (P=0.01). Next, we classified PCI scheduled patients (N=263) into groups with high, medium and low platelet densities per monocyte and assessed the recurrence of adverse events. After multivariate adjustment for potential confounders, we observed a 2.5-fold reduction in the recurrence of adverse events in patients with a high platelet density per monocyte as compared with a low platelet density per monocyte [hazard ratio=0.4 (95% confidence interval, 0.2-0.8), P=0.01]. We show that a high platelet density per monocyte increases monocyte recruitment to endothelial cells and predicts a reduction in the recurrence of adverse events in patients after PCI. These findings may imply that a high platelet density per monocyte protects against recurrence of adverse events.
Collapse
Affiliation(s)
- Bert Rutten
- Dr. Bert Rutten, Department of Clinical Chemistry and Haematology, UMC Utrecht, Utrecht, the Netherlands, E-mail:
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Loader J, Montero D, Lorenzen C, Watts R, Méziat C, Reboul C, Stewart S, Walther G. Acute Hyperglycemia Impairs Vascular Function in Healthy and Cardiometabolic Diseased Subjects. Arterioscler Thromb Vasc Biol 2015; 35:2060-72. [DOI: 10.1161/atvbaha.115.305530] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 06/16/2015] [Indexed: 02/04/2023]
Abstract
Objectives—
Controversy exists over the effect of acute hyperglycemia on vascular function. In this systematic review, we compared the effect of acute hyperglycemia on endothelial and vascular smooth muscle functions across healthy and cardiometabolic diseased subjects.
Approach and Results—
A systematic search of MEDLINE, EMBASE, and Web of Science from inception until July 2014 identified articles evaluating endothelial or vascular smooth muscle function during acute hyperglycemia and normoglycemia. Meta-analyses compared the standardized mean difference (SMD) in endothelial and vascular smooth muscle functions between acute hyperglycemia and normoglycemia. Subgroup analyses and metaregression identified sources of heterogeneity. Thirty-nine articles (525 healthy and 540 cardiometabolic subjects) were analyzed. Endothelial function was decreased (39 studies; n=1065; SMD, −1.25; 95% confidence interval, −1.52 to −0.98;
P
<0.01), whereas vascular smooth muscle function was preserved (6 studies; n=144; SMD, −0.07; 95% confidence interval, −0.30 to 0.16;
P
=0.55) during acute hyperglycemia compared with normoglycemia. Significant heterogeneity was detected among endothelial function studies (
P
<0.01). A subgroup analysis revealed that endothelial function was decreased in the macrocirculation (30 studies; n=884; SMD, −1.40; 95% confidence interval, −1.68 to −1.12;
P
<0.01) but not in the microcirculation (9 studies; n=181; SMD, −0.63; 95% confidence interval, −1.36 to 0.11;
P
=0.09). Similar results were observed according to health status. Macrovascular endothelial function was inversely associated with age, blood pressure, and low-density lipoprotein cholesterol and was positively associated with the postocclusion interval of vascular assessment.
Conclusions—
To our knowledge, this is the first systematic review and meta-analysis of its kind. In healthy and diseased subjects, we found evidence for macrovascular but not microvascular endothelial dysfunction during acute hyperglycemia.
Collapse
Affiliation(s)
- Jordan Loader
- From the Avignon University, LAPEC EA4278, Avignon, France (J.L., C.M., C.R., G.W.); School of Exercise Science (J.L., C.L., R.W., G.W.) and The Mary MacKillop Institute for Health Research (S.S.), Australian Catholic University, Melbourne, Victoria, Australia; and Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland (D.M.)
| | - David Montero
- From the Avignon University, LAPEC EA4278, Avignon, France (J.L., C.M., C.R., G.W.); School of Exercise Science (J.L., C.L., R.W., G.W.) and The Mary MacKillop Institute for Health Research (S.S.), Australian Catholic University, Melbourne, Victoria, Australia; and Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland (D.M.)
| | - Christian Lorenzen
- From the Avignon University, LAPEC EA4278, Avignon, France (J.L., C.M., C.R., G.W.); School of Exercise Science (J.L., C.L., R.W., G.W.) and The Mary MacKillop Institute for Health Research (S.S.), Australian Catholic University, Melbourne, Victoria, Australia; and Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland (D.M.)
| | - Rani Watts
- From the Avignon University, LAPEC EA4278, Avignon, France (J.L., C.M., C.R., G.W.); School of Exercise Science (J.L., C.L., R.W., G.W.) and The Mary MacKillop Institute for Health Research (S.S.), Australian Catholic University, Melbourne, Victoria, Australia; and Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland (D.M.)
| | - Cindy Méziat
- From the Avignon University, LAPEC EA4278, Avignon, France (J.L., C.M., C.R., G.W.); School of Exercise Science (J.L., C.L., R.W., G.W.) and The Mary MacKillop Institute for Health Research (S.S.), Australian Catholic University, Melbourne, Victoria, Australia; and Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland (D.M.)
| | - Cyril Reboul
- From the Avignon University, LAPEC EA4278, Avignon, France (J.L., C.M., C.R., G.W.); School of Exercise Science (J.L., C.L., R.W., G.W.) and The Mary MacKillop Institute for Health Research (S.S.), Australian Catholic University, Melbourne, Victoria, Australia; and Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland (D.M.)
| | - Simon Stewart
- From the Avignon University, LAPEC EA4278, Avignon, France (J.L., C.M., C.R., G.W.); School of Exercise Science (J.L., C.L., R.W., G.W.) and The Mary MacKillop Institute for Health Research (S.S.), Australian Catholic University, Melbourne, Victoria, Australia; and Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland (D.M.)
| | - Guillaume Walther
- From the Avignon University, LAPEC EA4278, Avignon, France (J.L., C.M., C.R., G.W.); School of Exercise Science (J.L., C.L., R.W., G.W.) and The Mary MacKillop Institute for Health Research (S.S.), Australian Catholic University, Melbourne, Victoria, Australia; and Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland (D.M.)
| |
Collapse
|
50
|
Lin S, Mequanint K. Activation of Transcription Factor GAX and Concomitant Downregulation of IL-1β and ERK1/2 Modulate Vascular Smooth Muscle Cell Phenotype in 3D Fibrous Scaffolds. Tissue Eng Part A 2015; 21:2356-65. [PMID: 26041434 DOI: 10.1089/ten.tea.2015.0153] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Since vascular smooth muscle cells (VSMCs) display phenotypic plasticity in response to changing environmental cues, understanding the molecular mechanisms underlying the phenotypic modulation mediated by a three-dimensional (3D) scaffold is important to engineer functional vasculature. Following cell seeding into 3D scaffolds, the synthetic phenotype is desired to enable cells to expand rapidly and produce and assemble extracellular matrix components, but must revert to a quiescent contractile phenotype after tissue fabrication to impart the contractile properties found in native blood vessels. This study shows that 3D electrospun fibrous scaffolds regulate human coronary artery smooth muscle cells (HCASMCs) toward a more synthetic phenotype characterized by reduced contractile markers, such as smooth muscle alpha-actin and calponin. The reduction in contractile markers expression was mediated by endogenously expressed proinflammatory cytokine interleukin-1β (IL-1β). 3D topography transiently induces concomitant upregulation of IL-1β and MAPK ERK1/2 through nuclear factor-κB-dependent signaling pathway. An early burst of expression of IL-1β is essential for suppression of the homeobox transcription factor Gax and related cyclin-dependent kinase inhibitor p21(Cip1), which are key regulators for cells exiting from cell cycle. Our findings provide new insights for understanding signaling mechanisms of HCASMCs in electrospun 3D fibrous scaffolds, which have considerable value for application in vascular tissue engineering.
Collapse
Affiliation(s)
- Shigang Lin
- 1 Department of Chemical and Biochemical Engineering, Faculty of Engineering, The University of Western Ontario , London, Canada
| | - Kibret Mequanint
- 1 Department of Chemical and Biochemical Engineering, Faculty of Engineering, The University of Western Ontario , London, Canada .,2 Graduate Program of Biomedical Engineering, The University of Western Ontario , London, Canada
| |
Collapse
|