1
|
Yu W, Kong Q, Jiang S, Li Y, Wang Z, Mao Q, Zhang X, Liu Q, Zhang P, Li Y, Li C, Ding Z, Liu L. HSPA12A maintains aerobic glycolytic homeostasis and Histone3 lactylation in cardiomyocytes to attenuate myocardial ischemia/reperfusion injury. JCI Insight 2024; 9:e169125. [PMID: 38421727 PMCID: PMC11128201 DOI: 10.1172/jci.insight.169125] [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: 01/26/2023] [Accepted: 02/21/2024] [Indexed: 03/02/2024] Open
Abstract
Myocardial ischemia/reperfusion (MI/R) injury is a major cause of adverse outcomes of revascularization following myocardial infarction. Anaerobic glycolysis during myocardial ischemia is well studied, but the role of aerobic glycolysis during the early phase of reperfusion is incompletely understood. Lactylation of Histone H3 (H3) is an epigenetic indicator of the glycolytic switch. Heat shock protein A12A (HSPA12A) is an atypic member of the HSP70 family. In the present study, we report that, during reperfusion following myocardial ischemia, HSPA12A was downregulated and aerobic glycolytic flux was decreased in cardiomyocytes. Notably, HSPA12A KO in mice exacerbated MI/R-induced aerobic glycolysis decrease, cardiomyocyte death, and cardiac dysfunction. Gain- and loss-of-function studies demonstrated that HSPA12A was required to support cardiomyocyte survival upon hypoxia/reoxygenation (H/R) challenge and that its protective effects were mediated by maintaining aerobic glycolytic homeostasis for H3 lactylation. Further analyses revealed that HSPA12A increased Smurf1-mediated Hif1α protein stability, thus increasing glycolytic gene expression to maintain appropriate aerobic glycolytic activity to sustain H3 lactylation during reperfusion and, ultimately, improving cardiomyocyte survival to attenuate MI/R injury.
Collapse
Affiliation(s)
- Wansu Yu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, and
| | - Qiuyue Kong
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Surong Jiang
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, and
| | - Yunfan Li
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhaohe Wang
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qian Mao
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaojin Zhang
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, and
| | - Qianhui Liu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, and
| | - Pengjun Zhang
- Department of Nuclear Medicine, Nanjing First Hospital of Nanjing Medical University, Nanjing, China
| | - Yuehua Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China
| | - Chuanfu Li
- Departments of Surgery, East Tennessee State University, Johnson City, Tennessee, USA
| | - Zhengnian Ding
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Li Liu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, and
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China
| |
Collapse
|
2
|
Xie Y, Wang Y, Pei W, Chen Y. Theranostic in GLP-1R molecular imaging: challenges and emerging opportunities. Front Mol Biosci 2023; 10:1210347. [PMID: 37780209 PMCID: PMC10540701 DOI: 10.3389/fmolb.2023.1210347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 09/06/2023] [Indexed: 10/03/2023] Open
Abstract
Theranostic in nuclear medicine combines diagnostic imaging and internal irradiation therapy using different therapeutic nuclear probes for visual diagnosis and precise treatment. GLP-1R is a popular receptor target in endocrine diseases, non-alcoholic steatohepatitis, tumors, and other areas. Likewise, it has also made breakthroughs in the development of molecular imaging. It was recognized that GLP-1R imaging originated from the study of insulinoma and afterwards was expanded in application including islet transplantation, pancreatic β-cell mass measurement, and ATP-dependent potassium channel-related endocrine diseases. Fortunately, GLP-1R molecular imaging has been involved in ischemic cardiomyocytes and neurodegenerative diseases. These signs illustrate the power of GLP-1R molecular imaging in the development of medicine. However, it is still limited to imaging diagnosis research in the current molecular imaging environment. The lack of molecular-targeted therapeutics related report hinders its radiology theranostic. In this article, the current research status, challenges, and emerging opportunities for GLP-1R molecular imaging are discussed in order to open a new path for theranostics and to promote the evolution of molecular medicine.
Collapse
Affiliation(s)
- Yang Xie
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, China
| | - Yudi Wang
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, China
| | - Wenjie Pei
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, China
| | - Yue Chen
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, China
| |
Collapse
|
3
|
Tang Y, Shi C, Qin Y, Wang S, Pan H, Chen M, Yu X, Lou Y, Fan G. Network Pharmacology-Based Investigation and Experimental Exploration of the Antiapoptotic Mechanism of Colchicine on Myocardial Ischemia Reperfusion Injury. Front Pharmacol 2022; 12:804030. [PMID: 34975499 PMCID: PMC8716846 DOI: 10.3389/fphar.2021.804030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022] Open
Abstract
Background: The beneficial effects of colchicine on cardiovascular disease have been widely reported in recent studies. Previous research demonstrated that colchicine has a certain protective effect on ischemic myocardium and has the potential to treat myocardial ischemia reperfusion injury (MIRI). However, the potential targets and pharmacological mechanism of colchicine to treat MIRI has not been reported. Methods: In this study, we used network pharmacology and experimental verification to investigate the pharmacological mechanisms of colchicine for the treatment of MIRI. Potential targets of colchicine and MIRI related genes were screened from public databases. The mechanism of colchicine in the treatment of MIRI was determined by protein-protein interaction (PPI), gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Additionally, we evaluated the effect of colchicine on H9C2 cell activity using CCK-8 assays, observed the effect of colchicine on H9C2 cell apoptosis via flow cytometry, and further verified the expression of key targets after colchicine treated by Western blot. Results: A total of 626 target genes for colchicine and 1549 MIRI disease targets were obtained. 138 overlapping genes were determined as potential targets of colchicine in treating MIRI. the PPI network analysis demonstrated that the targets linked to MIRI were ALB, TNF, ACTB, AKT1, IL6, TP53, IL1B, CASP3 and these targets showed nice affinity with colchicine in molecular docking experiments. The results of GO analysis and KEGG pathway enrichment demonstrated that the anti-MIRI effect of colchicine involves in apoptotic signaling pathway. Further tests suggested that colchicine can protect H9C2 cell from Hypoxia/Reoxygenation (H/R) injury through anti-apoptotic effects. Western blot results demonstrated that colchicine can inhibited MIRI induced apoptosis of H9C2 cell by enhancing the decreased levels of Caspase-3 in myocardial injure model induced by H/R and activating the PI3K/AKT/eNOS pathway. Conclusions: we performed network pharmacology and experimental evaluation to reveal the pharmacological mechanism of colchicine against MIRI. The results from this study could provide a theoretical basis for the development and clinical application of colchicine.
Collapse
Affiliation(s)
- Yuanjun Tang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chenyang Shi
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yingyi Qin
- Department of Health Statistics, Naval Medical University, Shanghai, China
| | - Shuowen Wang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hui Pan
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ming Chen
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xuemei Yu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuefen Lou
- Department of Pharmacy, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Guorong Fan
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Saraste A, Knuuti J. PET imaging in diabetic cardiomyopathy. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00051-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
5
|
Ma X, Liu Z, Ilyas I, Little PJ, Kamato D, Sahebka A, Chen Z, Luo S, Zheng X, Weng J, Xu S. GLP-1 receptor agonists (GLP-1RAs): cardiovascular actions and therapeutic potential. Int J Biol Sci 2021; 17:2050-2068. [PMID: 34131405 PMCID: PMC8193264 DOI: 10.7150/ijbs.59965] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/28/2021] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is closely associated with cardiovascular diseases (CVD), including atherosclerosis, hypertension and heart failure. Some anti-diabetic medications are linked with an increased risk of weight gain or hypoglycemia which may reduce the efficacy of the intended anti-hyperglycemic effects of these therapies. The recently developed receptor agonists for glucagon-like peptide-1 (GLP-1RAs), stimulate insulin secretion and reduce glycated hemoglobin levels without having side effects such as weight gain and hypoglycemia. In addition, GLP1-RAs demonstrate numerous cardiovascular protective effects in subjects with or without diabetes. There have been several cardiovascular outcomes trials (CVOTs) involving GLP-1RAs, which have supported the overall cardiovascular benefits of these drugs. GLP1-RAs lower plasma lipid levels and lower blood pressure (BP), both of which contribute to a reduction of atherosclerosis and reduced CVD. GLP-1R is expressed in multiple cardiovascular cell types such as monocyte/macrophages, smooth muscle cells, endothelial cells, and cardiomyocytes. Recent studies have indicated that the protective properties against endothelial dysfunction, anti-inflammatory effects on macrophages and the anti-proliferative action on smooth muscle cells may contribute to atheroprotection through GLP-1R signaling. In the present review, we describe the cardiovascular effects and underlying molecular mechanisms of action of GLP-1RAs in CVOTs, animal models and cultured cells, and address how these findings have transformed our understanding of the pharmacotherapy of T2DM and the prevention of CVD.
Collapse
Affiliation(s)
- Xiaoxuan Ma
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Zhenghong Liu
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Iqra Ilyas
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Peter J Little
- Sunshine Coast Health Institute, University of the Sunshine Coast, Birtinya, QLD 4575, Australia.,School of Pharmacy, Pharmacy Australia Centre of Excellence, the University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Danielle Kamato
- School of Pharmacy, Pharmacy Australia Centre of Excellence, the University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Amirhossein Sahebka
- Halal Research Center of IRI, FDA, Tehran, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad, Iran
| | - Zhengfang Chen
- Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu 215500, Jiangsu Province, China
| | - Sihui Luo
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Xueying Zheng
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Jianping Weng
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Suowen Xu
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| |
Collapse
|
6
|
Ståhle M, Kytö V, Kiugel M, Liljenbäck H, Metsälä O, Käkelä M, Li XG, Oikonen V, Saukko P, Nuutila P, Knuuti J, Roivainen A, Saraste A. Glucagon-like peptide-1 receptor expression after myocardial infarction: Imaging study using 68Ga-NODAGA-exendin-4 positron emission tomography. J Nucl Cardiol 2020; 27:2386-2397. [PMID: 30547299 PMCID: PMC7749060 DOI: 10.1007/s12350-018-01547-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Activation of glucagon-like peptide-1 receptor (GLP-1R) signaling protects against cardiac dysfunction and remodeling after myocardial infarction (MI). The aim of the study was to evaluate 68Ga-NODAGA-exendin-4 positron emission tomography (PET) for assessment of GLP-1R expression after MI in rats. METHODS AND RESULTS Rats were studied at 3 days, 1 and 12 weeks after permanent coronary ligation or a sham-operation. Rats were injected with 68Ga-NODAGA-exendin-4 and scanned with PET and contrast-enhanced computed tomography (CT) followed by digital autoradiography and histology of left ventricle tissue sections. 68Ga-NODAGA-exendin-4 PET/CT showed focally increased tracer uptake in the infarcted regions peaking at 3 days and continuing at 1 week after MI. Pre-treatment with an unlabeled exendin-4 peptide significantly reduced 68Ga-NODAGA-exendin-4 uptake. By autoradiography, 68Ga-NODAGA-exendin-4 uptake was 8.6-fold higher in the infarcted region and slightly increased also in the remote, non-infarcted myocardium at 1 week and 12 weeks post-MI compared with sham. Uptake of 68Ga-NODAGA-exendin-4 correlated with the amount of CD68-positive macrophages in the infarcted area and alpha-smooth muscle actin staining in the remote myocardium. CONCLUSIONS 68Ga-NODAGA-exendin-4 PET detects up-regulation of cardiac GLP-1R expression during healing of MI in rats and may provide information on the activated repair mechanisms after ischemic myocardial injury.
Collapse
Affiliation(s)
- Mia Ståhle
- Turku PET Centre, University of Turku, 20520 Turku, Finland
| | - Ville Kytö
- Heart Center, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Max Kiugel
- Turku PET Centre, University of Turku, 20520 Turku, Finland
| | - Heidi Liljenbäck
- Turku PET Centre, University of Turku, 20520 Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Olli Metsälä
- Turku PET Centre, University of Turku, 20520 Turku, Finland
| | - Meeri Käkelä
- Turku PET Centre, University of Turku, 20520 Turku, Finland
| | - Xiang-Guo Li
- Turku PET Centre, University of Turku, 20520 Turku, Finland
- Turku PET Centre, Åbo Akademi University, Turku, Finland
| | - Vesa Oikonen
- Turku PET Centre, University of Turku, 20520 Turku, Finland
| | - Pekka Saukko
- Department of Pathology and Forensic Medicine, University of Turku, Turku, Finland
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku, 20520 Turku, Finland
- Department of Endocrinology, Turku University Hospital, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Juhani Knuuti
- Turku PET Centre, University of Turku, 20520 Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, 20520 Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Antti Saraste
- Turku PET Centre, University of Turku, 20520 Turku, Finland
- Heart Center, Turku University Hospital, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
| |
Collapse
|
7
|
Malik N, Kornelsen R, McCormick S, Colpo N, Merkens H, Bendre S, Benard F, Sossi V, Schirrmacher R, Schaffer P. Development and biological evaluation of[ 18F]FMN3PA & [ 18F]FMN3PU for leucine-rich repeat kinase 2 (LRRK2) in vivo PET imaging. Eur J Med Chem 2020; 211:113005. [PMID: 33248850 DOI: 10.1016/j.ejmech.2020.113005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/22/2020] [Accepted: 11/07/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE Among all genetic mutations of LRRK2, the G2019S mutation is the most commonly associated with the late-onset of Parkinson's disease (PD). Hence, one potential therapeutic approach is to block the hyperactivity of mutated LRRK2 induced by kinase inhibition. To date, only a few LRRK2 kinase inhibitors have been tested for in vivo quantification of target engagement by positron emission tomography (PET). In this study, we performed biological evaluations of two radiolabeled kinase inhibitors i.e. [18F]FMN3PA (14) and [18F]FMN3PU for LRRK2 (15). PROCEDURES Radiosyntheses of [18F]FMN3PA (14) and [18F]FMN3PU (15) were performed using K[18F]-F-K222 complex in a TRACERlab FXN module and purification was carried out via C18 plus (Sep-Pak) cartridges. In vitro specific binding assays were performed in rat brain striatum and kidney tissues using GNE-0877 as a blocking agent (Ki = 0.7 nM). For in vivo blocking, 3 mg/kg of GNE-0877 was injected 30 min before radiotracer injection via tail vein in wild-type (WT) mice (n = 4). Dynamic scans by PET/CT (Siemens Inveon) were performed in WT mice (n = 3). RESULTS Radiofluorinations resulted in radiochemical yields (RCYs) of 25 ± 1.3% (n = 6) ([18F]FMN3PU, 15) and 37 ± 1.6% (n = 6) ([18F]FMN3PA, 14) with ≥96% radiochemical purity (RCP) and a molar activity (MA) of 3.55 ± 1.6 Ci/μmol (131 ± 56 GBq/μmol) for [18F]FMN3PU (15) and 4.57 ± 1.7 Ci/μmol (169 ± 63 GBq/μmol) for [18F]FMN3PA (14), respectively. Saturation assays showed high specific binding for rat brain striatum with Kd 20 ± 1.3 nM ([18F]FMN3PA, 14) and 23.6 ± 4.0 nM ([18F]FMN3PU, 15). In vivo blocking data for [18F]FMN3PA (14) was significant for brain (p < 0.0001, 77% blocking) and kidney (p = 0.0041, 65% blocking). PET images showed uptake in mouse brain striatum. CONCLUSION In the presence of GNE-0877 as a blocking agent, the specific binding of [18F]FMN3PA (14) and [18F]FMN3PU (15) was significant in vitro. [18F]FMN3PA (14) showed good brain uptake in vivo, though fast clearance from brain was observed (within 10-15 min).
Collapse
Affiliation(s)
| | | | | | - Nadine Colpo
- Molecular Oncology, British Columbia Cancer Research Institute, Canada
| | - Helen Merkens
- Molecular Oncology, British Columbia Cancer Research Institute, Canada
| | - Shreya Bendre
- Molecular Oncology, British Columbia Cancer Research Institute, Canada
| | - Francois Benard
- Molecular Oncology, British Columbia Cancer Research Institute, Canada; Department of Radiology, University of British Columbia, Canada
| | - Vesna Sossi
- Department of Physics and Astronomy, University of British Columbia, Canada
| | | | - Paul Schaffer
- Life Sciences Division, TRIUMF, Canada; Department of Radiology, University of British Columbia, Canada; Department of Chemistry, Simon Fraser University, Canada.
| |
Collapse
|
8
|
Zhang GJ, Wei LH, Lu HW, Xiao YF, Wang WR, He YL, Wang XM, Tian JH. Study on the mechanism of anti-MIRI action of total flavones of Fructus Chorspondiatis by PET/CT imaging. Cardiovasc Diagn Ther 2020; 10:796-810. [PMID: 32968635 DOI: 10.21037/cdt-20-305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background To investigate anti myocardial ischemia/reperfusion injury (MIRI) action of total flavones of Fructus Chorspondiatis (TFFC) in rats by 13N-ammonia micro PET/CT imaging, etc. Methods Male Sprague-Dawley rats were randomly divided into 6 groups. Micro PET/CT imaging was performed before and after modeling to calculate the volume (VOI) and SUVmean of myocardial ischemic area. The oxidative stress index [(superoxide dismutase (SOD), malondialdehyde (MDA)] and the marker enzymes [creatine kinase (CK), lactate dehydrogenase (LDH)] of myocardial injury were detected. The pathological changes of myocardial were observed via HE staining. A MIRI model of rat cardiomyocytes in vitro was established, the damage and apoptosis of myocardial cells in each group were observed, and the apoptosis rate of cardiomyocytes was detected. Results The imaging viscosities of the imaging agents were observed at 24 and 48 h in each group. The VOI of 24 h imaging was (6.33±2.02), (6.01±1.56) and (3.32±0.86) mm3, respectively. The VOI of 48 h imaging was (3.31±1.33), (2.61±1.01) and (1.32±0.58) mm3. The 72 h imaging medium and high dose group recovered, while the low dose group still saw sparseness with (1.26±0.68) mm3 VOI. The ischemic (SUVmean) gradually increased with time. Metabolism gradually recovered (F=121.82, 450.82, 435.75, P<0.05). The three doses of TFFC can eliminate free radicals and reduce the damage of myocardial injury. Amongst them, the high-dose group had a better effect on SOD, and the middle-dose group had a better effect on MDA and LDH. The low-dose group affected CK, and a significant difference was observed compared with the control group (P<0.05). After administration, the morphology of myocardial cells in each dose group was improved to some extent. Nuclear pyknosis, rupture, the apoptosis rate, etc. were significantly reduced, the number of cells increased. The high dose group showed the most obvious improvement. Conclusions The PET/CT imaging method can detect non-invasive, in vivo and dynamic MIRI, and can accurately evaluate the protective effect of traditional Mongolian medicine TFFC on MIRI. The Anti-MIRI of TFFC can scavenge free radicals, reduce oxidative stress damage, inhibit apoptosis, affect the activity of related enzymes.
Collapse
Affiliation(s)
- Guo-Jian Zhang
- Medical School of Chinese PLA, Beijing, China.,Department of Nuclear Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Li-Hong Wei
- Department of Pharmacy, Inner Mongolia International Mongolian Hospital, Hohhot, China
| | - Hai-Wen Lu
- Department of Imaging, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yun-Feng Xiao
- Center for New Drug Safety Evaluation and Research, Inner Mongolia Medical University, Hohhot, China
| | - Wen-Rui Wang
- Department of Nuclear Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yu-Lin He
- Department of Nuclear Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Xue-Mei Wang
- Department of Nuclear Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Jia-He Tian
- Medical School of Chinese PLA, Beijing, China
| |
Collapse
|
9
|
Velikyan I, Eriksson O. Advances in GLP-1 receptor targeting radiolabeled agent development and prospective of theranostics. Theranostics 2020; 10:437-461. [PMID: 31903131 PMCID: PMC6929622 DOI: 10.7150/thno.38366] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/10/2019] [Indexed: 12/15/2022] Open
Abstract
In the light of theranostics/radiotheranostics and prospective of personalized medicine in diabetes and oncology, this review presents prior and current advances in the development of radiolabeled imaging and radiotherapeutic exendin-based agents targeting glucagon-like peptide-1 receptor. The review covers chemistry, preclinical, and clinical evaluation. Such critical aspects as structure-activity-relationship, stability, physiological potency, kidney uptake, and dosimetry are discussed.
Collapse
Affiliation(s)
- Irina Velikyan
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Olof Eriksson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| |
Collapse
|
10
|
Pan X, Xu Q, Chen J, Wang T, Zhang M, Wang H, Gao H. Preliminary evaluation of 18F‑AlF‑NOTA‑MAL‑Cys40‑Exendin‑4 in rodent heart after myocardial ischemia and reperfusion. Mol Med Rep 2019; 20:2276-2284. [PMID: 31257516 PMCID: PMC6691274 DOI: 10.3892/mmr.2019.10432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 05/15/2019] [Indexed: 11/11/2022] Open
Abstract
Glucagon-like peptide-1 (GLP-1) and its receptor (GLP-1R) exert cardioprotective effects after myocardial ischemia and reperfusion (MI/R) in animal models and human clinical trials. Receptor imaging with positron emission tomography (PET) provides a non-invasive method for monitoring GLP-1R expression. In the present study, a fluorine-18-labeled aluminum fluoride exendin-4 analog [18F-AlF conjugated with 1,4,7-triazacyclononanetriacetic acid (NOTA)-maleimide (MAL)-Cys40-exendin-4] was synthesized and evaluated in a rat MI/R model for GLP-1R imaging. NOTA-MAL-Cys40-exendin-4 was synthesized by coupling Cys40-exendin-4 with NOTA-MAL. NOTA-MAL-Cys40-exendin-4 was then conjugated with 18F-AlF to obtain 18F-AlF-NOTA-MAL-Cys40-exendin-4. The yield of 18F-AlF-NOTA-MAL-Cys40-exendin-4 was 18.5±3.4% (not decay corrected). The process was completed within ~30 min. In rat MI/R models, the tracer exhibited specific binding to GLP-1R and an appropriate signal-to-noise ratio. At 8 h post-MI/R, tracer uptake reached its peak [0.35±0.053% of injected dose (%ID)/g; n=6] in ischemic myocardium. Localized tracer uptake decreased 1 day (0.20±0.032 %ID/g; n=6) and 3 days (0.16±0.017 %ID/g; n=6) post-MI/R compared with 8 h post-MI/R, but still remained higher compared with sham-operated groups (0.06±0.012 %ID/g; n=6). Pre-injected unlabeled exendin-4 effectively blocked tracer accumulation (0.09±0.041 %ID/g; n=6). In conclusion, 18F-AlF-NOTA-MAL-Cys40-exendin-4 demonstrated favorable characteristics for GLP-1R imaging following MI/R. PET imaging using 18F-AlF-NOTA-MAL-Cys40-exendin-4 in rodent hearts after MI/R revealed a dynamic pattern of GLP-1R upregulation.
Collapse
Affiliation(s)
- Xietian Pan
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Qing Xu
- Department of Radiation Oncology, The Affiliated Bayi Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210002, P.R. China
| | - Jiangwei Chen
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Tingting Wang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Mingming Zhang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Haichang Wang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Haokao Gao
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
11
|
Yue Y, Xu Y, Huang L, Pan D, Bai Z, Wang L, Yang R, Yan J, Song H, Li X, Yang M. Evaluation of A Novel GLP-1R Ligand for PET Imaging of Prostate Cancer. Anticancer Agents Med Chem 2018; 19:509-514. [PMID: 30068284 DOI: 10.2174/1871520618666180801101730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 07/20/2018] [Accepted: 07/21/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Glucagon-like peptide 1 receptor (GLP-1R) is an important biomarker for diagnosis and therapy of the endocrine cancers due to overexpression. Recently, in human prostate cancer cell lines the receptor was also observed, therefore it may be a potential target for the disease. 18F-Al-NOTA-MAL-Cys39- exendin-4 holds great promise for GLP-1R. Therefore, the feasibility of the 18F-labeled exendin-4 analog for prostate cancer imaging was investigated. METHODS New probe 18F-Al-NOTA-MAL-Cys39-exendin-4 was made through one-step fluorination. Prostate cancer PC3 cell xenograft model mice were established to primarily evaluate the imaging properties of the tracer via small animal PET studies in vivo. Pathological studies and Western Blots were also performed. RESULTS PC-3 prostate xenografts were clearly imaged under baseline conditions. At 30 and 60 min postinjection, the tumor uptakes were 2.90±0.41%ID/g and 2.26±0.32 %ID/g respectively. The presence of cys39-exendin-4 significantly reduced the tumor uptake to 0.82±0.10 %ID/g at 60 min p.i. Findings of ex vivo biodistribution studies were similar to those of in vivo PET imaging. The tumors to blood and muscles were significantly improved with the increase of time due to rapid clearance of the tracer from normal organs. Low levels of radioactivity were also detected in the GLP-1R positive tumor and normal organs after coinjection with excessive unlabeled peptides. Immunohistochemistry and Western Blots results confirmed that GLP-1R was widely expressed in PC-3 prostate cancers. CONCLUSION 18F-Al labeled exendin-4 analog might be a promising tracer for in vivo detecting GLP-1R positive prostate cancer with the advantage of facile synthesis and favorable pharmacokinetics. It may be useful in differential diagnosis, molecularly targeted therapy and prognosis of the cancers.
Collapse
Affiliation(s)
- Yuanyuan Yue
- Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Yuping Xu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| | - Lirong Huang
- Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Donghui Pan
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| | - Zhicheng Bai
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| | - Lizhen Wang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| | - Runlin Yang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| | - Junjie Yan
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| | - Huizhu Song
- Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Xiaotian Li
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450000, China
| | - Min Yang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, 214063, China
| |
Collapse
|
12
|
Automated radiosynthesis of [ 18F]FBEM, a sulfhydryl site specific labeling agent for peptides and proteins. Appl Radiat Isot 2018; 140:294-299. [PMID: 30098587 DOI: 10.1016/j.apradiso.2018.07.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 02/06/2023]
Abstract
In the process of developing [18F]FBEM coupled target peptide, we have instituted a robust automated synthesis of [18F]FBEM, a sulfhydryl (-SH) site specific agent for radiolabeling of peptides and proteins. The radiosynthesis generated 1.67-3.89 GBq (45.1-105.1 mCi, 7.5-18.8% non-decay corrected yield) of [18F]FBEM from 22.2 GBq (600 mCi) of starting [18F]fluoride with molar activity of 31.8 ± 5.3 GBq/µmol (0.86 ± 0.14 mCi/nmol) (n = 3) at the end of synthesis. Radiochemical purity was greater than 98%, and total synthesis time was ~90 min.
Collapse
|
13
|
Zhang J, Wu Z, Fan Z, Qin Z, Wang Y, Chen J, Wu M, Chen Y, Wu C, Wang J. Pericardial application as a new route for implanting stem-cell cardiospheres to treat myocardial infarction. J Physiol 2018; 596:2037-2054. [PMID: 29736937 PMCID: PMC5983168 DOI: 10.1113/jp275548] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/14/2018] [Indexed: 12/15/2022] Open
Abstract
KEY POINTS Cardiospheres (CSps) are a promising new form of cardiac stem cells with advantage over other stem cells for myocardial regeneration, but direct implantation of CSps by conventional routes has been limited due to potential embolism. We have implanted CSps into the pericardial cavity and systematically demonstrated its efficacy regarding myocardial infarction. Stem cell potency and cell viability can be optimized in vitro prior to implantation by pre-conditioning CSps with pericardial fluid and hydrogel packing. Transplantation of optimized CSps into the pericardial cavity improved cardiac function and alleviated myocardial fibrosis, increased myocardial cell survival and promoted angiogenesis. Mechanistically, CSps are able to directly differentiate into cardiomyocytes in vivo and promote regeneration of myocardial cells and blood vessels through a paracrine effect with released growth factors as potential paracrine mediators. These findings establish a new strategy for therapeutic myocardial regeneration to treat myocardial infarction. ABSTRACT Cardiospheres (CSps) are a new form of cardiac stem cells with an advantage over other stem cells for myocardial regeneration. However, direct implantation of CSps by conventional routes to treat myocardial infarction has been limited due to potential embolism. We have implanted CSps into the pericardial cavity and systematically assessed its efficacy on myocardial infarction. Preconditioning with pericardial fluid enhanced the activity of CSps and matrix hydrogel prolonged their viability. This shows that pretransplant optimization of stem cell potency and maintenance of cell viability can be achieved with CSps. Transplantation of optimized CSps into the pericardial cavity improved cardiac function and alleviated myocardial fibrosis in the non-infarcted area, and increased myocardial cell survival and promoted angiogenesis in the infarcted area. Mechanistically, CSps were able to directly differentiate into cardiomyocytes in vivo and promoted regeneration of myocardial cells and blood vessels in the infarcted area through a paracrine effect with released growth factors in pericardial cavity serving as possible paracrine mediators. This is the first demonstration of direct pericardial administration of pre-optimized CSps, and its effectiveness on myocardial infarction by functional and morphological outcomes with distinct mechanisms. These findings establish a new strategy for therapeutic myocardial regeneration to treat myocardial infarction.
Collapse
Affiliation(s)
- Jianhua Zhang
- Department of CardiologyThe Sun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangzhou510120PR China
- Department of CardiologyThe First Affiliated Hospital of Jinan UniversityGuangzhouPR China
| | - Zheng Wu
- Key Laboratory for Regenerative Medicine, Ministry of EducationJinan UniversityGuangzhouPR China
| | - Zepei Fan
- Key Laboratory for Regenerative Medicine, Ministry of EducationJinan UniversityGuangzhouPR China
| | - Zixi Qin
- Key Laboratory for Regenerative Medicine, Ministry of EducationJinan UniversityGuangzhouPR China
| | - Yingwei Wang
- Key Laboratory for Regenerative Medicine, Ministry of EducationJinan UniversityGuangzhouPR China
| | - Jiayuan Chen
- Department of CardiologyThe Sun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangzhou510120PR China
| | - Maoxiong Wu
- Department of CardiologyThe Sun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangzhou510120PR China
| | - Yangxin Chen
- Department of CardiologyThe Sun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangzhou510120PR China
| | - Changhao Wu
- Faculty of Health and Medical Sciences, School of Biosciences and MedicineUniversity of SurreyGuildfordGU2 7XHU.K.
| | - Jingfeng Wang
- Department of CardiologyThe Sun Yat‐sen Memorial Hospital of Sun Yat‐sen UniversityGuangzhou510120PR China
| |
Collapse
|
14
|
Deng H, Wang H, Zhang H, Wang M, Giglio B, Ma X, Jiang G, Yuan H, Wu Z, Li Z. Imaging Neurotensin Receptor in Prostate Cancer With 64Cu-Labeled Neurotensin Analogs. Mol Imaging 2018; 16:1536012117711369. [PMID: 28849698 PMCID: PMC6081756 DOI: 10.1177/1536012117711369] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
INTRODUCTION Neurotensin receptor 1 (NTR-1) is expressed and activated in prostate cancer cells. In this study, we explore the NTR expression in normal mouse tissues and study the positron emission tomography (PET) imaging of NTR in prostate cancer models. MATERIALS AND METHODS Three 64Cu chelators (1, 4, 7, 10-tetraazacyclododecane-1, 4, 7, 10-tetraacetic acid [DOTA], 1,4,7-triazacyclononane-N,N',N″-triacetic acid [NOTA], or AmBaSar) were conjugated to an NT analog. Neurotensin receptor binding affinity was evaluated using cell binding assay. The imaging profile of radiolabeled probes was compared in well-established NTR+ HT-29 tumor model. Stability of the probes was tested. The selected agents were further evaluated in human prostate cancer PC3 xenografts. RESULTS All 3 NT conjugates retained the majority of NTR binding affinity. In HT-29 tumor, all agents demonstrated prominent tumor uptake. Although comparable stability was observed, 64Cu-NOTA-NT and 64Cu-AmBaSar-NT demonstrated improved tumor to background contrast compared with 64Cu-DOTA-NT. Positron emission tomography/computed tomography imaging of the NTR expression in PC-3 xenografts showed high tumor uptake of the probes, correlating with the in vitro Western blot results. Blocking experiments further confirmed receptor specificity. CONCLUSIONS Our results demonstrated that 64Cu-labeled neurotensin analogs are promising imaging agents for NTR-positive tumors. These agents may help us identify NTR-positive lesions and predict which patients and individual tumors are likely to respond to novel interventions targeting NTR-1.
Collapse
Affiliation(s)
- Huaifu Deng
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,2 PET/CT Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hui Wang
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - He Zhang
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,3 Department of Radiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Mengzhe Wang
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ben Giglio
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xiaofen Ma
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,4 Department of Medical Imaging, Provincial People's Hospital, Guangzhou, China
| | - Guihua Jiang
- 4 Department of Medical Imaging, Provincial People's Hospital, Guangzhou, China
| | - Hong Yuan
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zhanhong Wu
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zibo Li
- 1 Department of Radiology, Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
15
|
Preparation of high bioactivity multilayered bone-marrow mesenchymal stem cell sheets for myocardial infarction using a 3D-dynamic system. Acta Biomater 2018; 72:182-195. [PMID: 29627677 DOI: 10.1016/j.actbio.2018.03.052] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 03/13/2018] [Accepted: 03/29/2018] [Indexed: 02/06/2023]
Abstract
Cell sheet techniques offer a promising future for myocardial infarction (MI) therapy; however, insufficient nutrition supply remains the major limitation in maintaining stem cell bioactivity in vitro. In order to enhance cell sheet mechanical strength and bioactivity, a decellularized porcine pericardium (DPP) scaffold was prepared by the phospholipase A2 method, and aspartic acid was used as a spacer arm to improve the vascular endothelial growth factor crosslink efficiency on the DPP scaffold. Based on this scaffold, multilayered bone marrow mesenchymal stem cell sheets were rapidly constructed, using RAD16-I peptide hydrogel as a temporary 3D scaffold, and cell sheets were cultured in either the 3D-dynamic system (DCcs) or the traditional static condition (SCcs). The multilayered structure, stem cell bioactivity, and ultrastructure of DCcs and SCcs were assessed. The DCcs exhibited lower apoptosis, lower differentiation, and an improved paracrine effect after a 48 h culture in vitro compared to the SCcs. Four groups were set to evaluate the cell sheet effect in rat MI model: sham group, MI control group, DCcs group, and SCcs group. The DCcs group improved cardiac function and decreased the infarcted area compared to the MI control group, while no significant improvements were observed in the SCcs group. Improved cell survival, angiogenesis, and Sca-1+ cell and c-kit+ cell amounts were observed in the DCcs group. In conclusion, the DCcs maintained higher stem cell bioactivity by using the 3D-dynamic system to provide sufficient nutrition, and transplanting DCcs significantly improved the cardiac function and angiogenesis. STATEMENT OF SIGNIFICANCE This study provides an efficient method to prepare vascular endothelial growth factor covalent decellularized pericardium scaffold with aspartic acid, and a multilayered bone marrow mesenchymal stem cell (BMSC) sheet is constructed on it using a 3D-dynamic system. The dynamic nutrition supply showed a significant benefit on BMSC bioactivity in vitro, including decreasing cell apoptosis, reducing stem cell differentiation, and improving growth factor secretion. These favorable bioactivity improved BMSC survival, angiogenesis, and cardiac function of the infarcted myocardium. The study highlights the importance of dynamic nutrition supply on maintaining stem cell bioactivity within cell sheet, and it stresses the necessity and significance of setting a standard for assessing cell sheet products before transplantation in the future application.
Collapse
|
16
|
Mechanisms of Cardiovascular Injury in Type 2 Diabetes and Potential Effects of Dipeptidyl Peptidase-4 Inhibition. J Cardiovasc Nurs 2017; 31:274-83. [PMID: 25829138 DOI: 10.1097/jcn.0000000000000245] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Cardiovascular (CV) disease is the major cause of mortality and morbidity in patients with type 2 diabetes mellitus (T2DM). The pathogenesis of CV disease in T2DM is complex and multifactorial and involves direct and indirect injury to the vasculature and heart. The impact of intensive glucose-lowering therapy with antihyperglycemic agents on CV outcomes is not clear, and questions remain as to which glucose-lowering agents may be beneficial to CV health in patients with T2DM. PURPOSE This review discusses findings regarding the known mechanisms of CV injury in T2DM and current knowledge regarding the potential cardioprotective effects of dipeptidyl peptidase-4 (DPP-4) inhibitors. CONCLUSIONS Dipeptidyl peptidase-4 inhibitors are relatively new antihyperglycemic agents. Their main mechanism of action is to inhibit the degradation of the incretin hormones glucagon-like peptide-1 and glucose-dependent insulinotropic peptide by DPP-4. By increasing levels of glucagon-like peptide-1, glucose-dependent insulin secretion is enhanced, glucagon secretion is suppressed, and the rate of gastric emptying is decreased. Dipeptidyl peptidase-4 also degrades other substances that are important in the regulation of CV function and inflammation. Animal studies, small observational studies in humans, and analyses of clinical trial data suggest that DPP-4 inhibitors may have beneficial CV effects. Recent prospectively designed CV outcomes trials with saxagliptin and alogliptin in patients with T2DM and high CV risk presented evidence that these DPP-4 inhibitors neither increased nor decreased adverse CV outcomes in this select patient population. CLINICAL IMPLICATIONS Dipeptidyl peptidase-4 inhibitors are promising therapies for the treatment of T2DM. Able to improve glycemic control without the risk of weight gain or hypoglycemia, they provide a safe alternative to sulfonylureas and are an effective adjunct to metformin. To date, this class of drugs seems to be at least neutral in terms of CV effects. Time will tell if these findings translate into a benefit for our patients.
Collapse
|
17
|
Zhong Z, Hu JQ, Wu XD, Sun Y, Jiang J. Anti-apoptotic effects of myocardin-related transcription factor-A on rat cardiomyocytes following hypoxia-induced injury. Can J Physiol Pharmacol 2016; 94:379-87. [PMID: 26854861 DOI: 10.1139/cjpp-2014-0461] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Myocardin-related transcription factor-A (MRTF-A) can transduce both biomechanical and humoral signals, which can positively modulate cardiac damage induced by acute myocardial infarction. However, the molecular mechanism that underlies the contribution that MRTF-A provides to the myocardium is not completely understood. The objective of this study was to investigate the effects of MRTF-A on myocardium apoptosis and its mechanisms. Our experiment results showed that MRTF-A expression increased and Bcl-2 expression reduced during myocardial ischemia–reperfusion in rat. Meanwhile, primary cardiomyocytes were pretreated with wild-type MRTF-A or siRNA of MRTF-A before exposure to hypoxia. We found that overexpression of MRTF-A in myocardial cells inhibited apoptosis and the release of cytochrome c. MRTF-A enhanced Bcl-2, which contributes to MRTF-A interaction with Bcl-2 in the nuclei of cardiomyocytes. MRTF-A upregulation expression of Bcl-2 in cardiomyocytes induced by hypoxia was inhibited by PD98059, an ERK1/2 inhibitor. In conclusions, MRTF-A improved myocardial cell survival in a cardiomyocyte model of hypoxia-induced injury; this effect was correlated with the upregulation of anti-apoptotic gene Bcl-2 through the activation of ERK1/2.
Collapse
Affiliation(s)
- Ze Zhong
- The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, JianDe 311600, China
| | - Jia-qing Hu
- The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, JianDe 311600, China
| | - Xin-dong Wu
- The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, JianDe 311600, China
| | - Yong Sun
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jun Jiang
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
18
|
Huang X, Zhang F, Sun X, Choi KY, Niu G, Zhang G, Guo J, Lee S, Chen X. The genotype-dependent influence of functionalized multiwalled carbon nanotubes on fetal development. Biomaterials 2014; 35:856-65. [PMID: 24344357 DOI: 10.1016/j.biomaterials.2013.10.027] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In many cases cancer is caused by gene deficiency that is being passed along from generation to generation. Soluble carbon nanotubes (CNTs) have shown promising applications in the diagnosis and therapy of cancer, however, the potential relationship between cancer-prone individuals and response to CNT exposure as a prerequisite for development of personalized nanomedicine, is still poorly understood. Here we report that intravenous injections of multi-walled carbon nanotubes into p53 (a well-known cancer-susceptible gene) heterozygous pregnant mice can induce p53- dependent responses in fetal development. Larger sized multi-walled carbon nanotubes moved across the blood-placenta barrier (BPB), restricted the development of fetuses, and induced brain deformity, whereas single-walled and smaller sized multi-walled carbon nanotubes showed no or less fetotoxicity. A molecular mechanism study found that multi-walled carbon nanotubes directly triggered p53-dependent apoptosis and cell cycle arrest in response to DNA damage. Based on the molecular mechanism, we also incorporated N-acetylcysteine (NAC), an FDA approved antioxidant, to prevent CNTs induced nuclear DNA damage and reduce brain development abnormalities. Our findings suggest that CNTs might have genetic background-dependent toxic effect on the normal development of the embryo, and provide new insights into protection against nanoparticle-induced toxicity in potential clinical applications.
Collapse
|
19
|
Yu Q, Fan W, Cao F. Mechanistic molecular imaging of cardiac cell therapy for ischemic heart disease. Am J Physiol Heart Circ Physiol 2013; 305:H947-59. [PMID: 23893164 DOI: 10.1152/ajpheart.00092.2013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cell-based myocardial regeneration has emerged as a promising therapeutic option for ischemic heart disease, though not yet at the level of routine clinical utility. Despite the encouraging results from initial preclinical studies that have demonstrated improved function and reduced infarct size of the ischemic myocardium following several candidate cell transplantation, the beneficial effects and molecular mechanisms of cardiac cell therapy are still unclear in clinical applications to date, and much remains to be optimized. To improve engraftment, accurate methods are required for tracking cell fate and quantifying functional outcome. In the present review, we summarized the current status and challenges of cardiac cell therapy for ischemic heart disease and discussed the strengths and limitations of currently available in vivo imaging techniques with special focus on the newly developed multimodality approaches for assessing the efficacy of engrafted donor cells. We also addressed the hurdles these imaging modalities are facing, including issues regarding immunogenicity and tumorigenicity of transplanted stem cells, and provided some the future perspectives on stem cell imaging.
Collapse
Affiliation(s)
- Qiujun Yu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | | |
Collapse
|
20
|
Kiesewetter DO, Guo N, Guo J, Gao H, Zhu L, Ma Y, Niu G, Chen X. Evaluation of an [(18)F]AlF-NOTA Analog of Exendin-4 for Imaging of GLP-1 Receptor in Insulinoma. Theranostics 2012; 2:999-1009. [PMID: 23139727 PMCID: PMC3493201 DOI: 10.7150/thno.5276] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Accepted: 10/16/2012] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION The GLP-1 receptor plays an important role in glucose homeostasis and thus is a very important target for diabetes therapy. The receptor is also overexpressed in insulinoma, a tumor of pancreatic beta-cells. We previously evaluated two fluorine-18-labeled analogs of exendin-4 prepared by conjugation with [(18)F]FBEM (N-[2-(4-[(18)F]fluorobenzamide)ethyl]maleimide). Both compounds demonstrated good tumor uptake, but the synthesis of the radiotracers was time consuming. To overcome this challenge, we developed a NOTA analog and performed radiolabeling using aluminum [(18)F]fluoride complexation. METHODS Cys(40)-exendin-4 was conjugated with NOTA mono N-ethylmaleimide. [(18)F]AlF conjugation was conducted and the radiolabeled product purified by preparative HPLC. Dynamic and static PET imaging scans were conducted on nude mice with established INS-1 xenografts. Uptake of tumor and other major organs in static images was quantitated (%ID/g) and comparison with blocking studies was made. PET quantification was also compared with ex vivo biodistribution results. RESULTS The radiosynthesis provided [(18)F]AlF-NOTA-MAL-cys(40)-exendin-4 in 23.6 ± 2.4 % radiochemical yield (uncorrected, n = 3) after HPLC; the process required about 55 min. The specific activity at time of injection ranged from 19.6 to 31.4 GBq (0.53-0.85 Ci)/µmol. Tumor uptake had reached its maximum (16.09 ± 1.18% ID/g, n = 4) by 5 min and remained nearly constant for the duration of the study. Kidney uptake continued to increase throughout the entire one hour time course. Pre-injection of exendin-4 caused a marked reduction in tissue uptake with the major exception of liver and kidneys, in which uptake was not affected. HPLC analysis of the radioactive components in extracts of the tumor and plasma showed primarily parent compound at 60 min post-injection, whereas extracts of kidney and urine contained exclusively one polar radioactive component. CONCLUSION The radiotracer is prepared in a simple one-step procedure and obtained in high specific activity after HPLC purification. [(18)F]AlF-NOTA-MAL-exendin-4 shows high tumor uptake and highly selective GLP-1 tissue uptake (INS-1 tumor, lung, pancreas), but still suffers from high kidney uptake.
Collapse
|