51
|
Xu J, Sun Y, Lu J. Knockdown of Long Noncoding RNA (lncRNA) AK094457 Relieved Angiotensin II Induced Vascular Endothelial Cell Injury. Med Sci Monit 2020; 26:e919854. [PMID: 32027625 PMCID: PMC7020760 DOI: 10.12659/msm.919854] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Hypertension could induce many serious diseases, including damage to vascular endothelial cells. As a non-coding RNA, long noncoding RNA (lncRNA) has received much attention in scientific research and has a regulating efficacy on many critical life activities in human body. The level of lncRNA AK094457 is thought to be elevated in hypertensive rats. However, there is no research indicating the relationship between the level of lncRNA AK094457 and vascular endothelial injury. Material/Methods In our study, we used lentiviral to knockdown lncRNA AK094457, and the human umbilical vein endothelial cells (HUVECs) were stimulated by the Ang II to imitate the vascular endothelial cell damage caused by hypertension. The Cell Counting Kit-8 assays were used to detect the cells viability. Western blotting was performed to detect the endothelial nitric oxide synthase (eNOS), p-eNOS and endothelin-1 (ET-1). After that the production of the NO was monitored. At last, the reactive oxygen species (ROS) levels and apoptosis rates were detected in this study. Results According to the results, we found that knockdown lncRNA AK094457 could alleviate the decrease of vascular endothelial cell viability induced by angiotensin II (Ang II). The knockdown of lncRNA AK094457 also relieved the downregulation of eNOS and p-eNOS, and the decreasing of NO release. At the same time, the knockdown of lncRNA inhibited the levels of Ang II-induced proinflammatory cytokines (tumor necrosis factor [TNF]-α, interleukin [IL]-1, and IL-6) and cell adhesion molecules (vascular cell adhesion molecule 1 [VCAM-1], intercellular adhesion molecule 1 [ICAM-1], and monocyte chemoattractant protein-1 [MCP-1]). The levels of ROS and apoptosis rates also decreased after the knockdown of lncRNA AK094457. Conclusions All these results indicated that lncRNA AK094457 could promote Ang II-induced vascular endothelial cell injury. On the contrary, knockdown of lncRNA AK094457 could alleviate this damage.
Collapse
Affiliation(s)
- JiaYi Xu
- Department of Gerontology, Minhang Hospital, Fudan University, Shanghai, China (mainland)
| | - Yingjie Sun
- Department of Critical Care Medicine, Minhang Hospital, Fudan University, Shanghai, China (mainland)
| | - Jie Lu
- Department of Gerontology, Minhang Hospital, Fudan University, Shanghai, China (mainland)
| |
Collapse
|
52
|
Laggner M, Gugerell A, Bachmann C, Hofbauer H, Vorstandlechner V, Seibold M, Gouya Lechner G, Peterbauer A, Madlener S, Demyanets S, Sorgenfrey D, Ostler T, Erb M, Mildner M, Ankersmit HJ. Reproducibility of GMP-compliant production of therapeutic stressed peripheral blood mononuclear cell-derived secretomes, a novel class of biological medicinal products. Stem Cell Res Ther 2020; 11:9. [PMID: 31900195 PMCID: PMC6942406 DOI: 10.1186/s13287-019-1524-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/29/2019] [Accepted: 12/10/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The recent concept of secretome-based tissue regeneration has profoundly altered the field of regenerative medicine and offers promising novel therapeutic options. In contrast to medicinal products with a single active substance, cell-derived secretomes comprise pleiotropic bioactive ingredients, representing a major obstacle for reproducible drug product efficacy and warranting patient safety. Good manufacturing practice (GMP)-compliant production guarantees high batch-to-batch consistency and reproducible efficacy of biological medicinal products, but different batches of cellular secretomes produced under GMP have not been compared yet, and suitable quality control parameters have not been established. To this end, we analyzed diverse biological and functional parameters of different batches produced under GMP of the secretome obtained from γ-irradiated peripheral blood mononuclear cells with proven tissue regenerative properties in infarcted myocardium, stroke, spinal cord injury, and skin wounds. METHODS We quantified key secretome ingredients, including cytokines, lipids, and extracellular vesicles, and functionally assessed potency in tube formation assay, ex vivo aortic ring sprouting assay, and cell-based protein and reporter gene assays. Furthermore, we determined secretome stability in different batches after 6 months of storage at various ambient temperatures. RESULTS We observed that inter-batch differences in the bioactive components and secretome properties were small despite considerable differences in protein concentrations and potencies between individual donor secretomes. Stability tests showed that the analytical and functional properties of the secretomes remained stable when lyophilisates were stored at temperatures up to + 5 °C for 6 months. CONCLUSIONS We are the first to demonstrate the consistent production of cell-derived, yet cell-free secretome as a biological medicinal product. The results from this study provide the basis for selecting appropriate quality control parameters for GMP-compliant production of therapeutic cell secretomes and pave the way for future clinical trials employing secretomes in tissue regenerative medicine.
Collapse
Affiliation(s)
- Maria Laggner
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
- Aposcience AG, Vienna, Austria
| | - Alfred Gugerell
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
- Aposcience AG, Vienna, Austria
| | | | - Helmut Hofbauer
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
- Aposcience AG, Vienna, Austria
| | - Vera Vorstandlechner
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
- Aposcience AG, Vienna, Austria
| | | | | | - Anja Peterbauer
- Austrian Red Cross Blood Transfusion Service of Upper Austria, Linz, Austria
| | - Sibylle Madlener
- Molecular Neuro-Oncology, Department of Pediatrics and Adolescent Medicine and Institute of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center of the Medical University of Vienna, Vienna, Austria
| | - Svitlana Demyanets
- Department for Laboratory Medicine at the Medical University of Vienna, Vienna, Austria
| | | | - Tobias Ostler
- SYNLAB Analytics and Services Switzerland AG, Birsfelden, Switzerland
| | - Michael Erb
- SYNLAB Analytics and Services Switzerland AG, Birsfelden, Switzerland
| | - Michael Mildner
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Hendrik Jan Ankersmit
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.
- Aposcience AG, Vienna, Austria.
| |
Collapse
|
53
|
Li Y, Wu P, Bihl JC, Shi H. Underlying Mechanisms and Potential Therapeutic Molecular Targets in Blood-Brain Barrier Disruption after Subarachnoid Hemorrhage. Curr Neuropharmacol 2020; 18:1168-1179. [PMID: 31903882 PMCID: PMC7770641 DOI: 10.2174/1570159x18666200106154203] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/18/2019] [Accepted: 01/04/2020] [Indexed: 01/01/2023] Open
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) is a subtype of hemorrhagic stroke with significant morbidity and mortality. Aneurysmal bleeding causes elevated intracranial pressure, decreased cerebral blood flow, global cerebral ischemia, brain edema, blood component extravasation, and accumulation of breakdown products. These post-SAH injuries can disrupt the integrity and function of the blood-brain barrier (BBB), and brain tissues are directly exposed to the neurotoxic blood contents and immune cells, which leads to secondary brain injuries including inflammation and oxidative stress, and other cascades. Though the exact mechanisms are not fully clarified, multiple interconnected and/or independent signaling pathways have been reported to be involved in BBB disruption after SAH. In addition, alleviation of BBB disruption through various pathways or chemicals has a neuroprotective effect on SAH. Hence, BBB permeability plays an important role in the pathological course and outcomes of SAH. This review discusses the recent understandings of the underlying mechanisms and potential therapeutic targets in BBB disruption after SAH, emphasizing the dysfunction of tight junctions and endothelial cells in the development of BBB disruption. The emerging molecular targets, including toll-like receptor 4, netrin-1, lipocalin-2, tropomyosin-related kinase receptor B, and receptor tyrosine kinase ErbB4, are also summarized in detail. Finally, we discussed the emerging treatments for BBB disruption after SAH and put forward our perspectives on future research.
Collapse
Affiliation(s)
| | | | - Ji C. Bihl
- Address correspondence to these authors at the Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio, 45435, USA; Tel: 011-01-9377755243; Fax: 011-01-9377757221; E-mail: and Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China; Tel: +86-15545107889; E-mail:
| | - Huaizhang Shi
- Address correspondence to these authors at the Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio, 45435, USA; Tel: 011-01-9377755243; Fax: 011-01-9377757221; E-mail: and Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China; Tel: +86-15545107889; E-mail:
| |
Collapse
|
54
|
Chen Q, Liu Y, Ding X, Li Q, Qiu F, Wang M, Shen Z, Zheng H, Fu G. Bone marrow mesenchymal stem cell-secreted exosomes carrying microRNA-125b protect against myocardial ischemia reperfusion injury via targeting SIRT7. Mol Cell Biochem 2019; 465:103-114. [PMID: 31858380 PMCID: PMC6955239 DOI: 10.1007/s11010-019-03671-z] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/30/2019] [Indexed: 12/15/2022]
Abstract
MicroRNA-125b (miR-125b) reduces myocardial infarct area and restrains myocardial ischemia reperfusion injury (I/R). In this study, we aimed to investigate the effect of bone marrow mesenchymal stem cell (BMSC)-derived exosomes carrying miR-125b on I/R rats. The myocardial I/R model in rats was constructed by ligation of the left anterior descending coronary artery (LAD). Rats were randomly divided into I/R and Sham group. Lv-cel-miR-67 (control) or Lv-miR-125b was transfected into BMSCs. Exosomes were extracted from transfected BMSCs, and separately named BMSC-Exo-67, BMSC-Exo-125b, and BMSC-Exo. MTT assay and flow cytometry were used to detect the viability and apoptosis of I/R myocardium cells, respectively. The expression of cell apoptosis proteins and the levels of inflammatory factors were examined by Western blot and ELISA assay, respectively. The target relationship between miR-125b and SIRT7 was predicted by using StarBase3.0, and was confirmed by using dual-luciferase reporter gene assay. qRT-PCR, immunohistochemistry staining, and Western blot were used to evaluate the expression of SIRT7 in myocardium tissues in I/R rats. BMSC-derived exosomes were successfully isolated and identified by TEM and positive expression of CD9 and CD63. The expression of miR-125b was down-regulated in I/R myocardium tissues and cells. BMSC-Exo-125b significantly up-regulated miR-125b in I/R myocardium cells. The intervention of BMSC-Exo-125b significantly increased the cell viability, decreased the apoptotic ratio, down-regulated Bax and caspase-3, up-regulated Bcl-2, and decreased the levels of IL-1β, IL-6, and TNF-α in I/R myocardium cells. SIRT7 was a target of miR-125b, and BMSC-Exo-125b significantly down-regulated SIRT7 in myocardium cells. In addition, the injection of BMSC-Exo-125b alleviated the pathological damages and down-regulated SIRT7 in myocardium tissues of I/R rats. BMSC-derived exosomes carrying miR-125b protected against myocardial I/R by targeting SIRT7.
Collapse
Affiliation(s)
- Qi Chen
- Department of Cardiology, School of Medicine, Sir Run Run Shaw Hospital, Biomedical Research Center, Zhejiang University, No. 3, East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Yu Liu
- Department of Cardiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, No. 321, Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Xueyan Ding
- Department of Cardiology, Zhejiang Provincial People's Hospital, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - Qinfeng Li
- Department of Cardiology, School of Medicine, Sir Run Run Shaw Hospital, Biomedical Research Center, Zhejiang University, No. 3, East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Fuyu Qiu
- Department of Cardiology, School of Medicine, Sir Run Run Shaw Hospital, Biomedical Research Center, Zhejiang University, No. 3, East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Meihui Wang
- Department of Cardiology, School of Medicine, Sir Run Run Shaw Hospital, Biomedical Research Center, Zhejiang University, No. 3, East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Zhida Shen
- Department of Cardiology, School of Medicine, Sir Run Run Shaw Hospital, Biomedical Research Center, Zhejiang University, No. 3, East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Hao Zheng
- Department of Cardiology, Zhejiang Provincial People's Hospital, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China.
| | - Guosheng Fu
- Department of Cardiology, School of Medicine, Sir Run Run Shaw Hospital, Biomedical Research Center, Zhejiang University, No. 3, East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
| |
Collapse
|
55
|
Extracellular HtrA2 Induces Apoptosis in Human Umbilical Vein Endothelial Cells. Int J Mol Sci 2019; 20:ijms20215446. [PMID: 31683713 PMCID: PMC6862081 DOI: 10.3390/ijms20215446] [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: 08/22/2019] [Revised: 10/17/2019] [Accepted: 10/26/2019] [Indexed: 12/11/2022] Open
Abstract
The serine protease high-temperature-required protein A2 (HtrA2) has been identified as a key intracellular molecule promoting apoptosis in cells during ischemia reperfusion (IR) injury. IR injury in ST-segment elevation myocardial infarction (STEMI) contributes to overall myocardial damage. HtrA2 has further been shown to be significantly increased in the serum of patients with STEMI. In the present pilot study, we use human umbilical vein endothelial cells (HUVECs) to investigate whether extracellular HtrA2 induces apoptosis using Annexin V staining. Furthermore, we examine whether HtrA2 is released extracellularly after staurosporine-induced apoptosis using ELISA. We find that HtrA2 is released upon induction of apoptosis by staurosporine into the cell culture medium. Furthermore, treatment of HUVECs with extracellular HtrA2-induces apoptosis, while the addition of anti-HtrA2 antibodies reduces both HtrA2- and staurosporine-induced endothelial cell apoptosis. In conclusion, we show here that extracellular HtrA2 induces apoptosis in human endothelial cells, although the exact molecular mechanisms have to be investigated in future.
Collapse
|
56
|
Kongpol K, Nernpermpisooth N, Prompunt E, Kumphune S. Endothelial-Cell-Derived Human Secretory Leukocyte Protease Inhibitor (SLPI) Protects Cardiomyocytes against Ischemia/Reperfusion Injury. Biomolecules 2019; 9:biom9110678. [PMID: 31683729 PMCID: PMC6920779 DOI: 10.3390/biom9110678] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/30/2019] [Accepted: 10/30/2019] [Indexed: 12/16/2022] Open
Abstract
Vascular endothelial cell (EC)-derived factors play an important role in endothelial-cardiomyocyte crosstalk and could save cardiomyocytes (CMs) from injury. The manipulation of endothelial cells to secrete protective factors could enhance cardioprotection. Secretory leukocyte protease inhibitor (SLPI) has been known to protect the heart. The goal of this study was to evaluate the in vitro paracrine protective effect and mechanisms of EC-derived human SLPI on cardiomyocytes subjected to hypoxia/reoxygenation (H/R) injury. Stable endothelial cells overexpressing human SLPI were generated from an endothelial cell line (EA.hy926). The cytoprotective effect was determined by cell survival assay. The results showed that endothelial-derived recombinant human SLPI (rhSLPI) reduced simulated ischemia/reperfusion (I/R)-(81.75% ± 1.42% vs. 60.27% ± 2.52%, p < 0.05) and hypoxia/reoxygenation (H/R)-induced EC injury (83.57% ± 1.78% vs. 63.07% ± 1.93%, p < 0.05). Moreover, co-culture of ECs overexpressing rhSLPI with CMs at ratios 1:1 and 1:3 or treatment with conditioned medium enhanced cell viability by 10.51-16.7% (co-culture) and 15.25-20.45% (conditioned medium) by reducing intracellular reactive oxygen species (ROS) production, the Bax/Bcl-2 expression ratio, caspase-3, and caspase-8, and in preconditioned CMs by activation of p38 MAPK and Akt survival kinase. In conclusion, this study showed for the first time that EC-derived rhSLPI provided cardio-vasculoprotective effects against I/R injury as a possible alternative therapeutic strategy for cardioprotection.
Collapse
Affiliation(s)
- Kantapich Kongpol
- Biomedical Research Unit in Cardiovascular Sciences (BRUCS), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand.
- Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand.
| | - Nitirut Nernpermpisooth
- Biomedical Research Unit in Cardiovascular Sciences (BRUCS), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand.
- Department of Cardio-Thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand.
| | - Eakkapote Prompunt
- Biomedical Research Unit in Cardiovascular Sciences (BRUCS), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand.
- Department of Medical Technology, School of Allied Health Sciences, University of Phayao, Phayao 56000, Thailand.
| | - Sarawut Kumphune
- Biomedical Research Unit in Cardiovascular Sciences (BRUCS), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand.
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand.
| |
Collapse
|
57
|
Li Y, Zhu X, Liu X, Du A, Yu B. miR-200a mediates protection of thymosin β-4 in cardiac microvascular endothelial cells as a novel mechanism under hypoxia-reoxygenation injury. J Cell Biochem 2019; 120:19098-19106. [PMID: 31265170 DOI: 10.1002/jcb.29237] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 08/22/2017] [Accepted: 08/24/2017] [Indexed: 12/29/2022]
Abstract
Thymosin β-4 (Tβ4) is a ubiquitous protein, which has been suggested to regulate multiple cell signal pathways and a variety of cellular functions. However, the role Tβ4 plays in the cardiac microvascular endothelial cells (CMECs) under myocardial ischemia/reperfusion injury is currently unknown. Here we investigated the effects of Tβ4 on hypoxia/reoxygenation (H/R) induced CMECs injury and its potential molecular mechanism. Cultured CMECs were positively identified by flow cytometry using antibody against CD31 and VWF/Factor VIII, which are constitutively expressed on the surface of CMECs. Then the reduced level of Tβ4 was detected in H/R-CMECs by a real-time quantitative polymerase chain reaction. To determine the effects of Tβ4 on H/R-CMECs, we transfected the overexpression or silence vector of Tβ4 into CMECs under H/R condition. Our results indicated that H/R treatment could reduce proliferation, increased apoptosis, adhesion, and reactive oxygen species (ROS) production in CMECs, which were attenuated by Tβ4 overexpression or aggravated by Tβ4 silencing, implying Tβ4 is able to promote CMECs against H/R-induced cell injury. Furthermore, the microRNA-200a (miR-200a) level was also increased by Tβ4 in H/R-CMECs or reduced by Tβ4 small interfering RNA. To investigated the mechanism of protective effects of Tβ4 on CMECs injury, the miR-200a inhibitor was transfected into H/R-CMECs. The results indicated that inhibition of miR-200a inversed the protection of Tβ4 on H/R-CMECs, specifically including cell proliferation, cell adhesion, cell apoptosis, and ROS production, as well as nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation. In conclusion, our results determined that Tβ4 attenuated H/R-induced CMECs injury by miR-200a-Nrf2 signaling.
Collapse
Affiliation(s)
- Yang Li
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Xiaolong Zhu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Xiping Liu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Aolin Du
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Bo Yu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| |
Collapse
|
58
|
Cardiac fibrosis: potential therapeutic targets. Transl Res 2019; 209:121-137. [PMID: 30930180 PMCID: PMC6545256 DOI: 10.1016/j.trsl.2019.03.001] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/01/2019] [Accepted: 03/05/2019] [Indexed: 01/14/2023]
Abstract
Cardiovascular disease is a leading cause of mortality in the world and is exacerbated by the presence of cardiac fibrosis, defined by the accumulation of noncontractile extracellular matrix proteins. Cardiac fibrosis is directly linked to cardiac dysfunction and increased risk of arrhythmia. Despite its prevalence, there is a lack of efficacious therapies for inhibiting or reversing cardiac fibrosis, largely due to the complexity of the cell types and signaling pathways involved. Ongoing research has aimed to understand the mechanisms of cardiac fibrosis and develop new therapies for treating scar formation. Major approaches include preventing the formation of scar tissue and replacing fibrous tissue with functional cardiomyocytes. While targeting the renin-angiotensin-aldosterone system is currently used as the standard line of therapy for heart failure, there has been increased interest in inhibiting the transforming growth factor-β signaling pathway due its established role in cardiac fibrosis. Significant advances in cell transplantation therapy and biomaterials engineering have also demonstrated potential in regenerating the myocardium. Novel techniques, such as cellular direct reprogramming, and molecular targets, such as noncoding RNAs and epigenetic modifiers, are uncovering novel therapeutic options targeting fibrosis. This review provides an overview of current approaches and discuss future directions for treating cardiac fibrosis.
Collapse
|
59
|
Differential effects of ischemia/reperfusion on endothelial function and contractility in donation after circulatory death. J Heart Lung Transplant 2019; 38:767-777. [DOI: 10.1016/j.healun.2019.03.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 02/21/2019] [Accepted: 03/08/2019] [Indexed: 01/18/2023] Open
|
60
|
Mi L, Zhang Y, Xu Y, Zheng X, Zhang X, Wang Z, Xue M, Jin X. HMGB1/RAGE pro-inflammatory axis promotes vascular endothelial cell apoptosis in limb ischemia/reperfusion injury. Biomed Pharmacother 2019; 116:109005. [PMID: 31136947 DOI: 10.1016/j.biopha.2019.109005] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE High-Mobility Group Box 1 (HMGB1) promotes vascular injuries induced by limb Ischemia and Reperfusion (IR), but the molecular mechanisms are not well understood. This study aimed to investigate the role of Receptor for Advanced-Glycation End products (RAGE) in HMGB1-regulated inflammatory response and vascular injury in limb IR using the rat IR and cellular Hypoxia and Reoxygenation (HR) models. METHODS We analyzed the vascular structure and elastic fiber deposition in rat femoral arteries by histological staining. We determined gene expression in vascular tissues and cells by quantitative RT-PCR, Western blotting and immunofluorescence; analyzed the protein levels in rat serum or cell supernatant by ELISA; and assessed protein interaction by co-immunoprecipitation. We used CCK-8 for analyzing cell viability, and assessed apoptosis by Hoechst staining and flow cytometry. RESULTS RAGE inhibition by FPS-ZM1 significantly repressed rat vascular injury that was induced by limb IR treatment. HMGB1 and RAGE expression, P38, ERK1/2, P65 and IKBa phosphorylation, as well as HIF-1a, NLRP3, Caspase-1, TNF-a and IL-6 expression and P65 in nucleus, increased in femoral arteries of a rat IR model and HUVEC undergoing HR treatment, whereas all the factors except HMGB1 and RAGE were inhibited by FPS-ZM1 treatment. In addition, we found that HMGB1 binds with RAGE in HUVEC undergoing HR treatment, which was suppressed by FPS-ZM1. Finally, the apoptosis of HUVEC also increased by HR treatment, but repressed under FPS-ZM1 treatment. CONCLUSION HMGB1 binds with RAGE to promote vascular inflammation and endothelial cell apoptosis, which mediates vascular injury during acute limb IR.
Collapse
Affiliation(s)
- Lei Mi
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China; Department of General Surgery, Taian City Central Hospital, Taian, Shandong, China
| | - Ying Zhang
- Department of Hepatobiliary Surgery, Taian City Central Hospital, Taian, Shandong, China
| | - Yugang Xu
- Department of General Surgery, Taian City Central Hospital, Taian, Shandong, China
| | - Xiao Zheng
- Department of General Surgery, Taian City Central Hospital, Taian, Shandong, China
| | - Xia Zhang
- Department of General Surgery, Taian City Central Hospital, Taian, Shandong, China
| | - Zhu Wang
- Department of Interventional Medicine and Vascular Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Ming Xue
- Department of Interventional Radiology, Weihai Municipal Hospital, Weihai, Shandong, China
| | - Xing Jin
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China.
| |
Collapse
|
61
|
Crouser ED, Matthay MA. Endothelial Damage During Septic Shock: Significance and Implications for Future Therapies. Chest 2019; 152:1-3. [PMID: 28693760 DOI: 10.1016/j.chest.2017.02.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 02/09/2017] [Indexed: 11/30/2022] Open
Affiliation(s)
- Elliott D Crouser
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, and the Dorothy M. Davis Heart and Lung Research Institute, Columbus, OH.
| | | |
Collapse
|
62
|
Feliu C, Peyret H, Poitevin G, Cazaubon Y, Oszust F, Nguyen P, Millart H, Djerada Z. Complementary Role of P2 and Adenosine Receptors in ATP Induced-Anti-Apoptotic Effects Against Hypoxic Injury of HUVECs. Int J Mol Sci 2019; 20:ijms20061446. [PMID: 30909368 PMCID: PMC6470483 DOI: 10.3390/ijms20061446] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/16/2019] [Accepted: 03/20/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Vascular endothelial injury during ischemia generates apoptotic cell death and precedes apoptosis of underlying tissues. We aimed at studying the role of extracellular adenosine triphosphate (ATP) on endothelial cells protection against hypoxia injury. METHODS In a hypoxic model on endothelial cells, we quantified the extracellular concentration of ATP and adenosine. The expression of mRNA (ectonucleotidases, adenosine, and P2 receptors) was measured. Apoptosis was evaluated by the expression of cleaved caspase 3. The involvement of P2 and adenosine receptors and signaling pathways was investigated using selective inhibitors. RESULTS Hypoxic stress induced a significant increase in extracellular ATP and adenosine. After a 2-h hypoxic injury, an increase of cleaved caspase 3 was observed. ATP anti-apoptotic effect was prevented by suramin, pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid (PPADS), and CGS15943, as well as by selective A2A, A2B, and A3 receptor antagonists. P2 receptor-mediated anti-apoptotic effect of ATP involved phosphoinositide 3-kinase (PI3K), extracellular signal-regulated kinases (ERK1/2), mitoKATP, and nitric oxide synthase (NOS) pathways whereas adenosine receptor-mediated anti-apoptotic effect involved ERK1/2, protein kinase A (PKA), and NOS. CONCLUSIONS These results suggest a complementary role of P2 and adenosine receptors in ATP-induced protective effects against hypoxia injury of endothelial. This could be considered therapeutic targets to limit the development of ischemic injury of organs such as heart, brain, and kidney.
Collapse
Affiliation(s)
- Catherine Feliu
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France.
| | - Hélène Peyret
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France.
| | - Gael Poitevin
- Laboratory of Hematology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France.
| | - Yoann Cazaubon
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France.
| | - Floriane Oszust
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France.
| | - Philippe Nguyen
- Laboratory of Hematology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France.
| | - Hervé Millart
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France.
| | - Zoubir Djerada
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France.
| |
Collapse
|
63
|
Dehaini H, Awada H, El-Yazbi A, Zouein FA, Issa K, Eid AA, Ibrahim M, Badran A, Baydoun E, Pintus G, Eid AH. MicroRNAs as Potential Pharmaco-targets in Ischemia-Reperfusion Injury Compounded by Diabetes. Cells 2019; 8:E152. [PMID: 30759843 PMCID: PMC6406262 DOI: 10.3390/cells8020152] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/06/2019] [Accepted: 02/10/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Ischemia-Reperfusion (I/R) injury is the tissue damage that results from re-oxygenation of ischemic tissues. There are many players that contribute to I/R injury. One of these factors is the family of microRNAs (miRNAs), which are currently being heavily studied. This review aims to critically summarize the latest papers that attributed roles of certain miRNAs in I/R injury, particularly in diabetic conditions and dissect their potential as novel pharmacologic targets in the treatment and management of diabetes. METHODS PubMed was searched for publications containing microRNA and I/R, in the absence or presence of diabetes. All papers that provided sufficient evidence linking miRNA with I/R, especially in the context of diabetes, were selected. Several miRNAs are found to be either pro-apoptotic, as in the case of miR-34a, miR-144, miR-155, and miR-200, or anti-apoptotic, as in the case of miR-210, miR-21, and miR-146a. Here, we further dissect the evidence that shows diverse cell-context dependent effects of these miRNAs, particularly in cardiomyocytes, endothelial, or leukocytes. We also provide insight into cases where the possibility of having two miRNAs working together to intensify a given response is noted. CONCLUSIONS This review arrives at the conclusion that the utilization of miRNAs as translational agents or pharmaco-targets in treating I/R injury in diabetic patients is promising and becoming increasingly clearer.
Collapse
Affiliation(s)
- Hassan Dehaini
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
| | - Hussein Awada
- Department of Biology, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
| | - Ahmed El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
- Department of Pharmacology and Toxicology, Alexandria University, Alexandria P.O. Box 21521, El-Mesallah, Egypt.
| | - Fouad A Zouein
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
| | - Khodr Issa
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
| | - Maryam Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
| | - Adnan Badran
- Department of Nutrition, University of Petra, Amman P.O Box 961343 Amman, Jordan.
| | - Elias Baydoun
- Department of Biology, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
| | - Gianfranco Pintus
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha P.O. Box 2713, Qatar.
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar.
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon.
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha P.O. Box 2713, Qatar.
| |
Collapse
|
64
|
Kanamaru H, Suzuki H. Potential therapeutic molecular targets for blood-brain barrier disruption after subarachnoid hemorrhage. Neural Regen Res 2019; 14:1138-1143. [PMID: 30804237 PMCID: PMC6425837 DOI: 10.4103/1673-5374.251190] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Aneurysmal subarachnoid hemorrhage remains serious hemorrhagic stroke with high morbidities and mortalities. Aneurysm rupture causes arterial bleeding-induced mechanical brain tissue injuries and elevated intracranial pressure, followed by global cerebral ischemia. Post-subarachnoid hemorrhage ischemia, tissue injuries as well as extravasated blood components and the breakdown products activate microglia, astrocytes and Toll-like receptor 4, and disrupt blood-brain barrier associated with the induction of many inflammatory and other cascades. Once blood-brain barrier is disrupted, brain tissues are directly exposed to harmful blood contents and immune cells, which aggravate brain injuries furthermore. Blood-brain barrier disruption after subarachnoid hemorrhage may be developed by a variety of mechanisms including endothelial cell apoptosis and disruption of tight junction proteins. Many molecules and pathways have been reported to disrupt the blood-brain barrier after subarachnoid hemorrhage, but the exact mechanisms remain unclear. Multiple independent and/or interconnected signaling pathways may be involved in blood-brain barrier disruption after subarachnoid hemorrhage. This review provides recent understandings of the mechanisms and the potential therapeutic targets of blood-brain barrier disruption after subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Hideki Kanamaru
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hidenori Suzuki
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
65
|
Wu Y, Zhang MH, Xue Y, Zhang T, Wu N, Guo W, Du X, Xu YL. Effect of microRNA-26a on vascular endothelial cell injury caused by lower extremity ischemia-reperfusion injury through the AMPK pathway by targeting PFKFB3. J Cell Physiol 2018; 234:2916-2928. [PMID: 30132885 DOI: 10.1002/jcp.27108] [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: 01/15/2018] [Accepted: 06/28/2018] [Indexed: 12/23/2022]
Abstract
Vascular endothelial cell (VEC) dysfunction plays an important role in the ischemia-reperfusion injury (IRI)-related diseases, and microRNAs (miRNAs) are key factors during this process. We conducted this study to investigate whether miRNA-26a (miR-26a) has effect on the IRI-induced VEC injury via the AMPK pathway by targeting 6-phosphofructo-2-kinase-fructose-2,6-biphosphatase 3 (PFKFB3). IRI rat models were successfully constructed by an abdominal incision. Additionally, the cultured VECs were further treated with miR-26a mimic or inhibitor, and si-PFKFB3. Both the reverse-transcription quantitative polymerase chain reaction and the western blot assay method were carried out to examine the expressions of PFKFB3, endothelial nitric oxide synthase (eNOS), and 5'-adenosine monophosphate-activated protein kinase (AMPK) α1, as well as the extent of the AMPK α1 phosphorylation levels in vascular tissues. Circulating endothelial cell (CEC), von Willebrand factor (VWF), thrombomodulin (TM), superoxide dismutase (SOD), malondialdehyde (MDA), nitric oxide (NO), and endothelin (ET) were all measured. In the rat model of an IRI, a poorly expressed miR-26a and contrarily highly expressed PFKFB3 were identified in vascular tissues. In response to an overexpression of miR-26a or to the PFKFB3 gene silencing, decreased CEC number, TM, VWF, MDA, and ET contents, increased AMPK α1, and eNOS levels, as well as the extent of AMPK α1 phosphorylation coordinate with both increased SOD and NO contents based on the restoration of the AMPK pathway. Overexpression of the miR-26a or si-PFKFB3 provides an elevation in cell proliferation. Our study suggests that the miR-26a RNA alleviates lower extremity IRI-induced VEC injury in rats through the activation of the AMPK pathway by inhibiting PFKFB3.
Collapse
Affiliation(s)
- Ye Wu
- Department of Vascular Surgery, Chinese PLA General Hospital, Beijing, China
| | - Min-Hong Zhang
- Department of Vascular Surgery, Chinese PLA General Hospital, Beijing, China
| | - Yan Xue
- Department of Vascular Surgery, Chinese PLA General Hospital, Beijing, China.,Department of Cardiovascular Surgery, General Hospital of Armed Police Forces, Beijing, China
| | - Tao Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, China
| | - Na Wu
- Department of Central Laboratory, Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing, China
| | - Wei Guo
- Department of Cardiovascular Surgery, Chinese PLA Rocket Force General Hospital, Beijing, China
| | - Xin Du
- Department of Vascular Surgery, Chinese PLA General Hospital, Beijing, China
| | - Yong-Le Xu
- Department of Vascular Surgery, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
66
|
Hernández-Reséndiz S, Muñoz-Vega M, Contreras WE, Crespo-Avilan GE, Rodriguez-Montesinos J, Arias-Carrión O, Pérez-Méndez O, Boisvert WA, Preissner KT, Cabrera-Fuentes HA. Responses of Endothelial Cells Towards Ischemic Conditioning Following Acute Myocardial Infarction. CONDITIONING MEDICINE 2018; 1:247-258. [PMID: 30338315 PMCID: PMC6191189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
One of the primary therapeutic goals of modern cardiology is to design strategies aimed at minimizing myocardial infarct size and optimizing cardiac function following acute myocardial infarction (AMI). Patients with AMI who underwent reperfusion therapy display dysfunction of the coronary endothelium. Consequently, ischemic endothelial cells become more permeable and weaken their natural anti-thrombotic and anti-inflammatory potential. Ischemia-reperfusion injury (IRI) is associated with activation of the humoral and cellular components of the hemostatic and innate immune system, and also with excessive production of reactive oxygen species (ROS), the inhibition of nitric oxide synthase, and with inflammatory processes. Given its essential role in the regulation of vascular homeostasis, involving platelets and leukocytes among others, dysfunctional endothelium can lead to increased risk of coronary vasospasm and thrombosis. Endothelial dysfunction can be prevented by ischemic conditioning with a protective intervention based on limited intermittent periods of ischemia and reperfusion. The molecular mechanisms and signal transduction pathways underlying conditioning phenomena in the coronary endothelium have been described as involving less ROS production, reduced adhesion of neutrophils to endothelial cells and diminished inflammatory reactions. This review summarizes our current understanding of the cellular and molecular mechanisms regulating IRI-affected and -damaged coronary endothelium, and how ischemic conditioning may preserve its function.
Collapse
Affiliation(s)
- Sauri Hernández-Reséndiz
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, NL, México
| | - Mónica Muñoz-Vega
- Unidad de Trastornos del Movimiento y Sueño, Hospital General Dr. Manuel Gea González. Ciudad de México, México
| | - Whendy E Contreras
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Gustavo E Crespo-Avilan
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | | | - Oscar Arias-Carrión
- Molecular Biology Department, Instituto Nacional de Cardiología "Ignacio Chávez", C.D de México, México
| | - Oscar Pérez-Méndez
- Molecular Biology Department, Instituto Nacional de Cardiología "Ignacio Chávez", C.D de México, México
| | - William A Boisvert
- Kazan Federal University, Department of Microbiology, Kazan, Russian Federation
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Hawaii, USA
| | - Klaus T Preissner
- Kazan Federal University, Department of Microbiology, Kazan, Russian Federation
- Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Hector A Cabrera-Fuentes
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, NL, México
- Kazan Federal University, Department of Microbiology, Kazan, Russian Federation
- Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
67
|
Endothelial Cell Dysfunction and Injury in Subarachnoid Hemorrhage. Mol Neurobiol 2018; 56:1992-2006. [DOI: 10.1007/s12035-018-1213-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 06/27/2018] [Indexed: 01/15/2023]
|
68
|
Calreticulin Ameliorates Hypoxia/Reoxygenation-Induced Human Microvascular Endothelial Cell Injury By Inhibiting Autophagy. Shock 2018; 49:108-116. [DOI: 10.1097/shk.0000000000000905] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
69
|
Nernpermpisooth N, Prompunt E, Kumphune S. An in vitro endothelial cell protective effect of secretory leukocyte protease inhibitor against simulated ischaemia/reperfusion injury. Exp Ther Med 2017; 14:5793-5800. [PMID: 29285123 PMCID: PMC5740774 DOI: 10.3892/etm.2017.5272] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 08/10/2017] [Indexed: 12/13/2022] Open
Abstract
Endothelial dysfunction is an essential deleterious modulator of ischaemia/reperfusion (I/R) injury. Secretory leukocyte protease inhibitor (SLPI) has demonstrated myocardial protection in cardiac transplantation; however, the effect of SLPI in endothelial I/R injury remains unexplored. In the present study, the effect of recombinant human SLPI (rhSLPI) treatment against endothelial cells (ECs) subjected to simulated I/R injury and the effect of treatment at different time points were determined. Human umbilical vein ECs (HUVECs) were subjected to normoxic or simulated I/R (sI/R) conditions, and rhSLPI at concentrations of 1, 10, 100 and 1,000 ng/ml was added to the cells prior to ischaemia, during ischaemia or at the onset of reperfusion. Endothelial injury and cytoskeleton disruption were assessed, and western blot analysis was conducted. The results revealed that rhSLPI treatment at 1,000 ng/ml significantly increased the HUVEC viability under sI/R injury (P<0.05). In addition, treatment with rhSLPI prior to or during ischaemia markedly attenuated the activity of lactase dehydrogenase compared with that in the sI/R group. In addition, the H2O2-induced reactive oxygen species production was reduced by ~17% upon rhSLPI pretreatment. Endothelial cytoskeleton disruption was also preserved by rhSLPI added prior to the reperfusion period. Furthermore, pretreatment with rhSLPI promoted protein kinase B activation, as well as reduced p38 mitogen-activated protein kinase phosphorylation and B-cell lymphoma 2-associated X protein expression in response to I/R injury. These findings indicated that rhSLPI possesses antioxidant and antiapoptotic properties against endothelial responses to I/R injury. Therefore, the cytoprotective effect of rhSLPI may provide a potential pharmaceutical target to limit endothelial-mediated I/R injury.
Collapse
Affiliation(s)
- Nitirut Nernpermpisooth
- Biomedical Research Unit in Cardiovascular Sciences (BRUCS), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand.,Department of Cardio-Thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Eakkapote Prompunt
- Biomedical Research Unit in Cardiovascular Sciences (BRUCS), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Sarawut Kumphune
- Biomedical Research Unit in Cardiovascular Sciences (BRUCS), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand.,Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| |
Collapse
|
70
|
Kristiansen SB, Sheykhzade M, Edvinsson L, Haanes KA. Changes in vasodilation following myocardial ischemia/reperfusion in rats. Nitric Oxide 2017; 70:68-75. [PMID: 28919322 DOI: 10.1016/j.niox.2017.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 08/01/2017] [Accepted: 09/12/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Blockage of a coronary artery, usually caused by arteriosclerosis, can lead to life threatening acute myocardial infarction. Opening with PCI (percutaneous coronary intervention), may be lifesaving, but reperfusion might exacerbate the cellular damage, and changes in the endothelium are believed to be involved in this worsened outcome. AIM The aim of the present study was to compare endothelial dependent and independent vasodilatory effect after experimental myocardial ischemia/reperfusion (I/R). METHODS A well-established rat model of myocardial ischemia with 24 h of reperfusion was applied, followed by a study in a wire myograph. RESULTS Endothelial NO dependent relaxation in response to carbachol, was sensitive to arterial depolarization, and was unaffected by I/R. In contrast, endothelial NO dependent ADPβS signalling, which was not sensitive to arterial depolarization, was significantly reduced after I/R. Following I/R, an H2O2 dependent EDH induced dilation appears in response to both of the above agonists. In addition, calcitonin gene-related peptide (CGRP) induced vasodilation was reduced. CONCLUSION These data show that NO dependent ADPβS induced dilation is reduced after I/R. However, there is some compensation by released H2O2 causing an EDH. Combined with a loss of maximal dilation in response to CGRP, the reduced vasodilation could be an important factor in understanding the exacerbated damage after I/R.
Collapse
Affiliation(s)
- Sarah Brøgger Kristiansen
- Department of Clinical Experimental Research, Glostrup Research Institute, Copenhagen University Hospital, Rigshospitalet-Glostrup, Denmark; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Majid Sheykhzade
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Lars Edvinsson
- Department of Clinical Experimental Research, Glostrup Research Institute, Copenhagen University Hospital, Rigshospitalet-Glostrup, Denmark
| | - Kristian Agmund Haanes
- Department of Clinical Experimental Research, Glostrup Research Institute, Copenhagen University Hospital, Rigshospitalet-Glostrup, Denmark.
| |
Collapse
|
71
|
Akbar N, Digby JE, Cahill TJ, Tavare AN, Corbin AL, Saluja S, Dawkins S, Edgar L, Rawlings N, Ziberna K, McNeill E, Johnson E, Aljabali AA, Dragovic RA, Rohling M, Belgard TG, Udalova IA, Greaves DR, Channon KM, Riley PR, Anthony DC, Choudhury RP. Endothelium-derived extracellular vesicles promote splenic monocyte mobilization in myocardial infarction. JCI Insight 2017; 2:93344. [PMID: 28878126 PMCID: PMC5621885 DOI: 10.1172/jci.insight.93344] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 07/27/2017] [Indexed: 12/31/2022] Open
Abstract
Transcriptionally activated monocytes are recruited to the heart after acute myocardial infarction (AMI). After AMI in mice and humans, the number of extracellular vesicles (EVs) increased acutely. In humans, EV number correlated closely with the extent of myocardial injury. We hypothesized that EVs mediate splenic monocyte mobilization and program transcription following AMI. Some plasma EVs bear endothelial cell (EC) integrins, and both proinflammatory stimulation of ECs and AMI significantly increased VCAM-1-positive EV release. Injected EC-EVs localized to the spleen and interacted with, and mobilized, splenic monocytes in otherwise naive, healthy animals. Analysis of human plasma EV-associated miRNA showed 12 markedly enriched miRNAs after AMI; functional enrichment analyses identified 1,869 putative mRNA targets, which regulate relevant cellular functions (e.g., proliferation and cell movement). Furthermore, gene ontology termed positive chemotaxis as the most enriched pathway for the miRNA-mRNA targets. Among the identified EV miRNAs, EC-associated miRNA-126-3p and -5p were highly regulated after AMI. miRNA-126-3p and -5p regulate cell adhesion- and chemotaxis-associated genes, including the negative regulator of cell motility, plexin-B2. EC-EV exposure significantly downregulated plexin-B2 mRNA in monocytes and upregulated motility integrin ITGB2. These findings identify EVs as a possible novel signaling pathway by linking ischemic myocardium with monocyte mobilization and transcriptional activation following AMI.
Collapse
Affiliation(s)
- Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Janet E. Digby
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Thomas J. Cahill
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Abhijeet N. Tavare
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Alastair L. Corbin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Sushant Saluja
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Sam Dawkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Laurienne Edgar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Nadiia Rawlings
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Klemen Ziberna
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Eileen McNeill
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | | | | | - Alaa A. Aljabali
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | | | - Mala Rohling
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | | | - Irina A. Udalova
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | | | - Keith M. Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Paul R. Riley
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | | | - Robin P. Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
- Acute Vascular Imaging Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
72
|
Cai H, Yao Z, Li W. IRF-5 accelerates leukocyte adhesion to endothelial cells in ischemia-reperfusion injury through regulating the transcription of VCAM-1. Biochem Biophys Res Commun 2017; 492:192-198. [PMID: 28818665 DOI: 10.1016/j.bbrc.2017.08.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 08/12/2017] [Indexed: 10/19/2022]
Abstract
Ischemia-reperfusion injury (IRI) has been implicated in many pathological conditions, including cardiovascular diseases. Adhesion of leukocytes to the surface of endothelial cells has been considered as one of the principle steps in the pathological cascade of inflammatory tissue damage during IRI. The role of the transcriptional factor interferon regulatory factor-5 (IRF-5) in endothelial physiology remains unknown. Here, we report that IRF-5 is expressed in human umbilical vein endothelial cells (HUVECs) and is rapidly upregulated in response to IRI, mediated by the JAK2/STAT3 pathway. Importantly, IRF-5 is involved in IRI-induced attachment of THP-1 leukocytes to HUVECs. Mechanistically, it was found that IRF-5 targeted the expression of vascular cell adhesion molecule 1 (VCAM-1) at the transcriptional level by binding to its promoter. In conclusion, we identify IRF-5 as a new regulator and thus a therapeutic target in IRI-driven cardiovascular pathologies.
Collapse
Affiliation(s)
- Hongbin Cai
- Department of Cardiology, Tianjin People's Hospital, Tianjin, 300120, China
| | - Zhuhua Yao
- Department of Cardiology, Tianjin People's Hospital, Tianjin, 300120, China.
| | - Wenting Li
- Department of Cardiology, Tianjin People's Hospital, Tianjin, 300120, China
| |
Collapse
|
73
|
Vieceli Dalla Sega F, Aquila G, Fortini F, Vaccarezza M, Secchiero P, Rizzo P, Campo G. Context-dependent function of ROS in the vascular endothelium: The role of the Notch pathway and shear stress. Biofactors 2017; 43:475-485. [PMID: 28419584 DOI: 10.1002/biof.1359] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/12/2017] [Indexed: 12/15/2022]
Abstract
Reactive oxygen species (ROS) act as signal molecules in several biological processes whereas excessive, unregulated, ROS production contributes to the development of pathological conditions including endothelial dysfunction and atherosclerosis. The maintenance of a healthy endothelium depends on many factors and on their reciprocal interactions; in this framework, the Notch pathway and shear stress (SS) play two lead roles. Recently, evidence of a crosstalk between ROS, Notch, and SS, is emerging. The aim of this review is to describe the way ROS interact with the Notch pathway and SS protecting from-or promoting-the development of endothelial dysfunction. © 2017 BioFactors, 43(4):475-485, 2017.
Collapse
Affiliation(s)
| | - Giorgio Aquila
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Francesca Fortini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Mauro Vaccarezza
- Faculty of Health Sciences, School of Biomedical Sciences, Curtin University, Perth, Australia
| | - Paola Secchiero
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA) Center, Ferrara, Italy
| | - Paola Rizzo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
- Maria Cecilia Hospital, GVM Care & Research, E.S. Health Science Foundation, Cotignola, (RA), Italy
| | - Gianluca Campo
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria S. Anna, Cona, (FE), Italy
| |
Collapse
|
74
|
Wong BW, Marsch E, Treps L, Baes M, Carmeliet P. Endothelial cell metabolism in health and disease: impact of hypoxia. EMBO J 2017. [PMID: 28637793 DOI: 10.15252/embj.201696150] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In contrast to the general belief, endothelial cell (EC) metabolism has recently been identified as a driver rather than a bystander effect of angiogenesis in health and disease. Indeed, different EC subtypes present with distinct metabolic properties, which determine their function in angiogenesis upon growth factor stimulation. One of the main stimulators of angiogenesis is hypoxia, frequently observed in disease settings such as cancer and atherosclerosis. It has long been established that hypoxic signalling and metabolism changes are highly interlinked. In this review, we will provide an overview of the literature and recent findings on hypoxia-driven EC function and metabolism in health and disease. We summarize evidence on metabolic crosstalk between different hypoxic cell types with ECs and suggest new metabolic targets.
Collapse
Affiliation(s)
- Brian W Wong
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Leuven Cancer Institute KU Leuven, Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Elke Marsch
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Leuven Cancer Institute KU Leuven, Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Leuven Cancer Institute KU Leuven, Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Myriam Baes
- Laboratory for Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Leuven Cancer Institute KU Leuven, Leuven, Belgium .,Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| |
Collapse
|
75
|
García-Ruiz JM, Galán-Arriola C, Fernández-Jiménez R, Aguero J, Sánchez-González J, García-Alvarez A, Nuno-Ayala M, Dubé GP, Zafirelis Z, López-Martín GJ, Bernal JA, Lara-Pezzi E, Fuster V, Ibáñez B. Bloodless reperfusion with the oxygen carrier HBOC-201 in acute myocardial infarction: a novel platform for cardioprotective probes delivery. Basic Res Cardiol 2017; 112:17. [PMID: 28188434 DOI: 10.1007/s00395-017-0605-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 01/23/2017] [Indexed: 02/08/2023]
Abstract
Reperfusion, despite being required for myocardial salvage, is associated with additional injury. We hypothesize that infarct size (IS) will be reduced by a period of bloodless reperfusion with hemoglobin-based oxygen carriers (HBOC) before blood-flow restoration. In the pig model, we first characterized the impact of intracoronary perfusion with a fixed volume (600 ml) of a pre-oxygenated acellular HBOC, HBOC-201, on the healthy myocardium. HBOC-201 was administered through the lumen of the angioplasty balloon (i.e., distal to the occlusion site) immediately after onset of coronary occlusion at 1, 0.7, 0.4, or 0.2 ml/kg/min for 12, 17, 30, and 60 min, respectively, followed by blood-flow restoration. Outcome measures were systemic hemodynamics and LV performance assessed by the state-of-the-art cardiac magnetic resonance (CMR) imaging. The best performing HBOC-201 perfusion strategies were then tested for their impact on LV performance during myocardial infarction, in pigs subjected to 45 min mid-left anterior descending (LAD) coronary occlusion. At the end of the ischemia duration, pigs were randomized to regular reperfusion (blood-only reperfusion) vs. bloodless reperfusion (perfusion with pre-oxygenated HBOC-201 distal to the occlusion site), followed by blood-flow restoration. Hemodynamics and CMR-measured LV performance were assessed at 7- and 45-day follow-up. In modifications of the HBOC-201 procedure, glucose and insulin were included to support cardiac metabolism. A total of 66 pigs were included in this study. Twenty healthy pigs (5 per infusion protocol) were used in the study of healthy myocardium. Intracoronary administration of HBOC-201 (600 ml) at varying rates, including a flow of 0.4 ml/kg/min (corresponding to a maximum perfusion time of 30 min), did not damage the healthy myocardium. Slower perfusion (longer infusion time) was associated with permanent LV dysfunction and myocardial necrosis. A total of 46 pigs underwent MI induction. Compared with regular reperfusion, bloodless reperfusion with pre-oxygenated HBOC-201 alone increased IS. This effect was reversed by enrichment of pre-oxygenated HBOC-201 solution with glucose and insulin, resulting in no increase in IS or worsening of long-term ventricular function despite further delaying restoration of blood flow in the LAD. Bloodless reperfusion with a pre-oxygenated HBOC-201 solution supplemented with glucose and insulin is feasible and safe, but did not reduce infarct size. This strategy could be, however, used to deliver agents to the myocardium to treat or prevent ischemia/reperfusion injury before blood-flow restoration.
Collapse
Affiliation(s)
- Jose M García-Ruiz
- Myocardial Pathophysiology Area, Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.,CIBER de Enfermedades CardioVasculares (CIBERCV), Madrid, Spain.,Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Carlos Galán-Arriola
- Myocardial Pathophysiology Area, Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.,CIBER de Enfermedades CardioVasculares (CIBERCV), Madrid, Spain
| | - Rodrigo Fernández-Jiménez
- Myocardial Pathophysiology Area, Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.,CIBER de Enfermedades CardioVasculares (CIBERCV), Madrid, Spain.,The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jaume Aguero
- Myocardial Pathophysiology Area, Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.,CIBER de Enfermedades CardioVasculares (CIBERCV), Madrid, Spain
| | | | - Ana García-Alvarez
- Myocardial Pathophysiology Area, Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.,Hospital Clinic, Barcelona, Spain
| | - Mario Nuno-Ayala
- Myocardial Pathophysiology Area, Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | | | | | - Gonzalo J López-Martín
- Myocardial Pathophysiology Area, Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Juan A Bernal
- Myocardial Pathophysiology Area, Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Enrique Lara-Pezzi
- Myocardial Pathophysiology Area, Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.,CIBER de Enfermedades CardioVasculares (CIBERCV), Madrid, Spain
| | - Valentín Fuster
- Myocardial Pathophysiology Area, Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.,The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Borja Ibáñez
- Myocardial Pathophysiology Area, Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain. .,CIBER de Enfermedades CardioVasculares (CIBERCV), Madrid, Spain. .,Department of Cardiology, Instituto de Investigación Sanitaria, Fundación Jiménez Díaz, Madrid, Spain.
| |
Collapse
|
76
|
Unacylated ghrelin analog prevents myocardial reperfusion injury independently of permeability transition pore. Basic Res Cardiol 2016; 112:4. [PMID: 27995363 DOI: 10.1007/s00395-016-0595-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 12/09/2016] [Indexed: 02/06/2023]
Abstract
Reperfusion injury is responsible for an important part of myocardial infarct establishment due notably to triggering cardiomyocytes death at the first minutes of reperfusion. AZP-531 is an optimized analog of unacylated ghrelin currently in clinical development in several metabolic diseases. We investigated a potential cardioprotective effect of AZP-531 in ischemia/reperfusion (IR) and the molecular underlying mechanism(s) involved in this protection. In vivo postconditioning with AZP-531 in C57BL6 mouse IR model decreased infarct size. Western blot analysis on areas at risk from the different mouse groups showed that AZP-531 activates Akt, ERK1-2 as well as S6 and 4EBP1, mTORC1 effectors. We also showed an inhibition of caspase 3 cleavage and Bax translocation to the mitochondria. AZP-531 also stimulated the expression of antioxidants and was capable of decreasing mitochondrial H2O2 production, contributing to the reduction of ROS accumulation. AZP-531 exhibits cardioprotective effect when administrated for postconditioning in C57BL6 mouse IR model. Treatment with AZP-531 rescued the myocardium from cell death at early reperfusion by stimulating protein synthesis, inhibiting Bax/caspase 3-induced apoptosis as well as ROS accumulation and oxidative stress-induced necrosis. AZP-531 may prove useful in the treatment of IR injury.
Collapse
|
77
|
Cui H, Li N, Li X, Qi K, Li Q, Jin C, Wang T, Duan L, Jiang L, Chen G, Wang Z, Wei C, Yang Y. Tongxinluo modulates cytokine secretion by cardiac microvascular endothelial cells in ischemia/reperfusion injury. Am J Transl Res 2016; 8:4370-4381. [PMID: 27830021 PMCID: PMC5095330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 09/26/2016] [Indexed: 06/06/2023]
Abstract
Cardiac microvascular endothelial cells (CMECs) extensively secrete cytokines during myocardial ischemia/reperfusion injury (MIRI). Tongxinluo (TXL) has been demonstrated to preserve the function of the endothelium and myocardium against MIRI. This study was designed to identify alterations in the paracrine function of CMECs under hypoxia/reoxygenation (H/R) conditions and assess its modulation by TXL. CMECs were exposed to different concentrations of TXL for 30 min and then subjected to hypoxia and reoxygenation for 12 and 2 h, respectively. Apoptosis was measured to determine the optimal TXL concentration. Protein antibody arrays were used to assess changes in cytokines secreted into conditioned medium by CMECs. A Gene Ontology (GO) analysis was applied to interpret the functional implications of changes in cytokines. TXL inhibited CMEC apoptosis in a concentration-dependent manner after H/R, reaching peak efficacy at a concentration of 800 μg/ml. H/R significantly altered 33 cytokines, and TXL (800 μg/ml) changed the levels of 121 different cytokines compared with the H/R group. Among these cytokines, 10 that were increased by H/R were decreased by TXL, five that were decreased by H/R were increased by TXL, and eight that were attenuated by H/R were further decreased by TXL. Insulin-like growth factor binding protein-1 was up-regulated by H/R and was further increased by TXL. Significantly altered factors were found to be involved in cell proliferation, growth and differentiation, as well as chemotaxis and transport. TXL inhibited the apoptosis of CMECs and modulated their paracrine function in MIRI.
Collapse
Affiliation(s)
- Hehe Cui
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Na Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Xiangdong Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Kang Qi
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Qing Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Chen Jin
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Tianjie Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Lian Duan
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Leipei Jiang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Guihao Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Zhigang Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Cong Wei
- The Integration of Traditional and Western Medical Research Academy of Hebei ProvinceShijiazhuang, Hebei, China
| | - Yuejin Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| |
Collapse
|
78
|
Kong Q, Dai L, Wang Y, Zhang X, Li C, Jiang S, Li Y, Ding Z, Liu L. HSPA12B Attenuated Acute Myocardial Ischemia/reperfusion Injury via Maintaining Endothelial Integrity in a PI3K/Akt/mTOR-dependent Mechanism. Sci Rep 2016; 6:33636. [PMID: 27644317 PMCID: PMC5028890 DOI: 10.1038/srep33636] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/31/2016] [Indexed: 12/22/2022] Open
Abstract
Endothelial damage is a critical mediator of myocardial ischemia/reperfusion (I/R) injury. HSPA12B is an endothelial-cell-specifically expressed heat shock protein. However, the roles of HSPA12B in acute myocardial I/R injury is unknown. Here we reported that myocardial I/R upregulated HSPA12B expression in ventricular tissues, and endothelial overexpression of HSPA12B in transgenic mice (Tg) limited infarct size, attenuated cardiac dysfunction and improved cardiomyocyte survival compared with their wild type littermates. These improvements were accompanied with the diminished myocardial no-reflow phenomenon, decreased microvascular leakage, and better maintained endothelial tight junctions. The I/R-evoked neutrophil infiltration was also suppressed in Tg hearts compared with its wild type (WT) littermates. Moreover, Tg hearts exhibited the enhanced activation of PI3K/Akt//mTOR signaling following I/R challenge. However, pharmacological inhibition of PI3K abolished the HSPA12B-induced cardioprotection against myocardial I/R injury. The data demonstrate for the first time that the endothelial HSPA12B protected hearts against myocardial I/R injury. This cardioprotective action of HSPA12B was mediated, at least in part, by improving endothelial integrity in a PI3K/Akt/mTOR-dependent mechanism. Our study suggests that targeting endothelial HSPA12B could be an alternative approach for the management of patients with myocardial I/R injury.
Collapse
Affiliation(s)
- Qiuyue Kong
- Department of Anesthesiology, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Leyang Dai
- Department of Anesthesiology, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Yana Wang
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Xiaojin Zhang
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Chuanfu Li
- Departments of Surgery, East Tennessee State University, Johnson City, TN37614, USA
| | - Surong Jiang
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Yuehua Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing 210029, China
| | - Zhengnian Ding
- Department of Anesthesiology, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Li Liu
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
79
|
Kim BS, Jacobs D, Emontzpohl C, Goetzenich A, Soppert J, Jarchow M, Schindler L, Averdunk L, Kraemer S, Marx G, Bernhagen J, Pallua N, Schlemmer HP, Simons D, Stoppe C. Myocardial Ischemia Induces SDF-1α Release in Cardiac Surgery Patients. J Cardiovasc Transl Res 2016; 9:230-238. [PMID: 27055858 DOI: 10.1007/s12265-016-9689-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/22/2016] [Indexed: 01/07/2023]
Abstract
In the present observational study, we measured serum levels of the chemokine stromal cell-derived factor-1α (SDF-1α) in 100 patients undergoing cardiac surgery with cardiopulmonary bypass at seven distinct time points including preoperative values, myocardial ischemia, reperfusion, and the postoperative course. Myocardial ischemia triggered a marked increase of SDF-1α serum levels whereas cardiac reperfusion had no significant influence. Perioperative SDF-1α serum levels were influenced by patients' characteristics (e.g., age, gender, aspirin intake). In an explorative analysis, we observed an inverse association between SDF-1α serum levels and the incidence of organ dysfunction. In conclusion, time of myocardial ischemia was identified as the key stimulus for a significant upregulation of SDF-1α, indicating its role as a marker of myocardial injury. The inverse association between SDF-1α levels and organ dysfunction association encourages further studies to evaluate its organoprotective properties in cardiac surgery patients.
Collapse
Affiliation(s)
- Bong-Sung Kim
- Department of Plastic Surgery, Hand Surgery, Burn Center, RWTH Aachen University, Aachen, Germany
| | - Denise Jacobs
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Christoph Emontzpohl
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Andreas Goetzenich
- Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany
| | - Josefin Soppert
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Mareike Jarchow
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Lisa Schindler
- Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany.,Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany
| | - Luisa Averdunk
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Sandra Kraemer
- Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany
| | - Gernot Marx
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Jürgen Bernhagen
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Norbert Pallua
- Department of Plastic Surgery, Hand Surgery, Burn Center, RWTH Aachen University, Aachen, Germany
| | | | - David Simons
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Christian Stoppe
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany.
| |
Collapse
|
80
|
Jose Corbalan J, Vatner DE, Vatner SF. Myocardial apoptosis in heart disease: does the emperor have clothes? Basic Res Cardiol 2016; 111:31. [PMID: 27043720 DOI: 10.1007/s00395-016-0549-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 03/24/2016] [Indexed: 01/06/2023]
Abstract
Since the discovery of a novel mechanism of cell death that differs from traditional necrosis, i.e., apoptosis, there have been numerous studies concluding that increased apoptosis augments myocardial infarction and heart failure and that limiting apoptosis protects the heart. Importantly, the vast majority of cells in the heart are non-myocytes with only roughly 30 % myocytes, yet almost the entire field studying apoptosis in the heart has disregarded non-myocyte apoptosis, e.g., only 4.7 % of 423 studies on myocardial apoptosis in the past 3 years quantified non-myocyte apoptosis. Accordingly, we reviewed the history of apoptosis in the heart focusing first on myocyte apoptosis, followed by the history of non-myocyte apoptosis in myocardial infarction and heart failure. Apoptosis of several of the major non-myocyte cell types in the heart (cardiac fibroblasts, endothelial cells, vascular smooth muscle cells, macrophages and leukocytes) may actually be responsible for affecting the severity of myocardial infarction and heart failure. In summary, even though it is now known that the majority of apoptosis in the heart occurs in non-myocytes, very little work has been done to elucidate the mechanisms by which non-myocyte apoptosis might be responsible for the adverse effects of apoptosis in myocardial infarction and heart failure. The goal of this review is to provide an impetus for future work in this field on non-myocyte apoptosis that will be required for a better understanding of the role of apoptosis in the heart.
Collapse
Affiliation(s)
- J Jose Corbalan
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Dorothy E Vatner
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Stephen F Vatner
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA.
| |
Collapse
|
81
|
Liu RR, Li J, Gong JY, Kuang F, Liu JY, Zhang YS, Ma QL, Song CJ, Truax AD, Gao F, Yang K, Jin BQ, Chen LH. MicroRNA-141 regulates the expression level of ICAM-1 on endothelium to decrease myocardial ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol 2015; 309:H1303-13. [PMID: 26371161 DOI: 10.1152/ajpheart.00290.2015] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 09/02/2015] [Indexed: 01/01/2023]
Abstract
A growing number of studies have suggested microRNAs (miRNAs) are involved in the modulation of myocardial ischemia-reperfusion (MI/R) injury; however, the role of endogenous miRNAs targeting endothelial cells (ECs) and its interaction with ICAM-1 in the setting of MI/R remain poorly understood. Our microarray results showed that miR-146a, miR-146b-5p, miR-155*, miR-155, miR-497, and miR-451 were significantly upregulated, whereas, miR-141 and miR-564 were significantly downregulated in the ECs challenged with TNF-α for 6 h. Real-time PCR analyses additionally validated that the expression levels of miR-146a, miR-155*, and miR-141 were consistent with the microarray results. Then, ICAM-1 was identified as a novel target of miR-141 by Target Scan software and the reporter gene system. Further functional experiments showed that elevated levels of miR-141 inhibited ICAM-1 expression and diminished leukocytes adhesion to ECs in vitro. In an in vivo murine model of MI/R injury, pretreatment with miR-141 mimics through the tail vein downregulated the expression level of ICAM-1 in heart and attenuated MI/R injury as evidenced by decreased infarct size and decline of serum cardial troponin I (cTnI) and lactate dehydrogenase (LDH) concentration. The cardioprotective effects of miR-141 mimics may be attributed to the decreased infiltration of CD11b(+) cells and F4/80(+) macrophages into ischemic myocardium tissue. In conclusion, our results demonstrate that miR-141, as a novel repressor of ICAM-1, is involved in the attenuation of MI/R injury via antithetical regulation of ICAM-1 and inflammatory cells infiltration. Thus miR-141 may constitute a new therapeutic target in the setting of ischemic heart disease.
Collapse
Affiliation(s)
- Rong Rong Liu
- Department of Immunology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jun Li
- Department of Physiology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jiu Yu Gong
- Department of Immunology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Fang Kuang
- Department of Neurobiology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jia Yun Liu
- Department of Clinical Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China; and
| | - Yu Si Zhang
- Department of Immunology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Qian Li Ma
- Department of Immunology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Chao Jun Song
- Department of Immunology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Agnieszka D Truax
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Feng Gao
- Department of Physiology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Kun Yang
- Department of Immunology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Bo Quan Jin
- Department of Immunology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Li Hua Chen
- Department of Immunology, Fourth Military Medical University, Xi'an, People's Republic of China;
| |
Collapse
|
82
|
Obesity induced by high fat diet attenuates postinfarct myocardial remodeling and dysfunction in adult B6D2F1 mice. J Mol Cell Cardiol 2015; 84:154-61. [PMID: 25953257 DOI: 10.1016/j.yjmcc.2015.04.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/27/2015] [Accepted: 04/28/2015] [Indexed: 01/09/2023]
Abstract
Obesity is a major risk factor for cardiovascular morbidity and mortality. However, some studies suggest that among patients with established cardiovascular disease, obesity is associated with better prognosis, a phenomenon described as the obesity paradox. In this study we tested the hypothesis that obesity with hyperinsulinemia and without hyperglycemia attenuates the impact of transient coronary occlusion on left ventricular remodeling and function. B6D2F1 mice from both genders fed with a high fat diet (HFD) or control diet for 6 months were subjected to 45 min of coronary occlusion and 28 days of reperfusion. Left ventricular dimensions and function were assessed by serial echocardiography, and infarct size was determined by Picrosirius red staining. HFD mice developed obesity with hypercholesterolemia and hyperinsulinemia in the absence of hyperglycemia or hypertension. During the period of feeding, no changes were observed in ventricular mass, volume or function, or in vascular reactivity. HFD attenuated the consequences of transient coronary occlusion as shown by a marked reduction in infarct size (51%, P = 0.021) and cardiac dilation, as well as improved left ventricular function as compared to control diet animals. These effects were associated with enhanced reperfusion injury salvage kinases (RISK) pathway function in HFD hearts shown as increased Akt and GSK3β phosphorylation. These results demonstrate that dietary obesity without hyperglycemia or hypertension attenuates the impact of ischemia/reperfusion injury in association with increased insulin signaling and RISK activation. This study provides experimental support to the controversial concept of the obesity paradox in humans.
Collapse
|
83
|
Induction of autophagy by Tongxinluo through the MEK/ERK pathway protects human cardiac microvascular endothelial cells from hypoxia/reoxygenation injury. J Cardiovasc Pharmacol 2015; 64:180-90. [PMID: 24705173 DOI: 10.1097/fjc.0000000000000104] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
: In contrast to cardiomyocytes, autophagy in cardiac microvascular endothelial cells (CMECs) during ischemia/reperfusion (I/R) injury has not been fully investigated. Tongxinluo (TXL), a traditional Chinese medicine, was shown to be vascular protective. We aimed to elucidate the role of autophagy and its regulatory mechanisms by TXL in CMECs subjected to I/R injury. CMECs were exposed to different treatments for 30 minutes and subjected to hypoxia/reoxygenation each for 2 hours. The results indicated that hypoxia/reoxygenation significantly induced autophagy, as identified by an increased number of monodansylcadaverine-positive CMECs, increased autophagosome formation, and a higher type II/type I of light chain 3 ratio, but not Beclin-1 expression. Autophagy inhibition using 3-methyladenine was proapoptotic, but rapamycin-induced autophagy was antiapoptotic. TXL enhanced autophagy and decreased apoptosis in a dose-dependent manner, reaching its largest effect at 800 μg/mL. 3-methyladenine attenuated the TXL-promoted autophagy and antiapoptotic effects, whereas rapamycin had no additional effects compared with TXL alone. TXL upregulated mitogen-activated protein kinase and extracellular signal-regulated kinase (ERK) phosphorylation; however, PD98059 abrogated ERK phosphorylation and decreased autophagy and increased apoptosis compared with TXL alone. These results suggest that autophagy is a protective mechanism in CMECs subjected to I/R injury and that TXL can promote autophagy through activation of the mitogen-activated protein kinase/ERK pathway.
Collapse
|
84
|
Endothelial Insulin Resistance Protects the Heart Against Prolonged Ischemia–Reperfusion Injury But Does Not Prevent Insulin Transport Across the Endothelium in a Mouse Langendorff Model. J Cardiovasc Pharmacol Ther 2014; 19:586-91. [DOI: 10.1177/1074248414525506] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Aim: The endothelium plays an important role in the maintenance of cardiovascular homeostasis in healthy individuals. Insulin resistance can lead to the development of endothelial dysfunction, which is an important step in the pathogenesis of atherosclerosis. We investigated specifically whether the presence of vascular insulin resistance and endothelial dysfunction has any influence on the myocardial tolerance to ischemia–reperfusion (IR) injury, using Endothelial Specific Mutant Insulin Receptor Over-expressing (ESMIRO) mice, which exhibit vascular insulin resistance and vascular dysfunction. Methods: ESMIRO or wild-type (WT) littermate mouse hearts were isolated and perfused on a Langendorff apparatus. These were subjected to either 35-minute or 45-minute ischemia followed by reperfusion, after which infarct size was determined. The ability of insulin to activate its target kinase pathway, that is, phosphoinositide 3 (PI3) kinase/protein kinase B (AKT) in ESMIRO hearts was also assessed by Western blot analysis. Results: Compared to 35-minute ischemia, the extended 45-minute ischemic protocol significantly exacerbated myocardial infarction in WT mice, (56% ± 4%, n = 6 vs 32% ± 4%, n = 9; P < .01) but not in ESMIRO littermates (34% ± 7%, n = 6 vs 32% ± 3%, n = 9; not significant), suggesting some form of protective phenotype. Insulin treatment was associated with a significant increase in AKT phosphorylation in the myocardium in both the ESMIRO mice and WT littermates, and this was attenuated in both by inhibition of PI3 kinase using LY294002. Thus, insulin was able to directly activate PI3 kinase/AKT in the myocardium despite the absence of functional endothelial insulin receptors in the ESMIRO mice. Conclusion: (1) Insulin at pharmacologic concentrations can be transported across the endothelium independent of vascular insulin receptors and (2) vascular insulin resistance and/or endothelial dysfunction are protective against prolonged IR injury in the Langendorff model.
Collapse
|
85
|
Wu Z, Lu X, Zhou X, Chen Z, Zhao Y. The role of inflammation in postresuscitation myocardial dysfunction. Int J Cardiol 2013; 169:e47-8. [PMID: 24054316 DOI: 10.1016/j.ijcard.2013.08.124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Accepted: 08/29/2013] [Indexed: 10/26/2022]
Affiliation(s)
- Zhe Wu
- Medical College of Wuhan University, 9 Zhangzhidong Road, Wuhan 430060, PR China
| | | | | | | | | |
Collapse
|
86
|
Chen S, Synowsky S, Tinti M, MacKintosh C. The capture of phosphoproteins by 14-3-3 proteins mediates actions of insulin. Trends Endocrinol Metab 2011; 22:429-36. [PMID: 21871813 DOI: 10.1016/j.tem.2011.07.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Revised: 06/26/2011] [Accepted: 07/25/2011] [Indexed: 10/17/2022]
Abstract
How does signalling via PI3K-PKB (AKT)-mTORC1-p70S6K and ERK-p90RSK mediate wide-ranging physiological responses to insulin? Quantitative proteomics and biochemical experiments are revealing that these signalling pathways induce the phosphorylation of large and overlapping sets of proteins, which are then captured by phosphoprotein-binding proteins named 14-3-3s. The 14-3-3s are dimers that dock onto dual-phosphorylated sites in a configuration with special signalling and mechanical properties. They interact with the Rab GTPase-activating proteins AS160 and TBC1D1 to regulate glucose uptake into target tissues in response to insulin and energy stress. Dynamic patterns in the 14-3-3-binding phosphoproteome are providing new insights into how insulin triggers coherent shifts in metabolism that are integrated with other cellular response systems.
Collapse
Affiliation(s)
- Shuai Chen
- MRC Protein Phosphorylation Unit, College of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | | | | | | |
Collapse
|
87
|
Badalzadeh R, Mohammadi M, Najafi M, Ahmadiasl N, Farajnia S, Ebrahimi H. The additive effects of ischemic postconditioning and cyclosporine-A on nitric oxide activity and functions of diabetic myocardium injured by ischemia/reperfusion. J Cardiovasc Pharmacol Ther 2011; 17:181-9. [PMID: 21828282 DOI: 10.1177/1074248411416118] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The interaction of diabetes with cardioprotection by postconditioning in ischemia/reperfusion injury remains unclear. The aim of this study was to investigate the concomitant effects of ischemic postconditioning (IPostC) and cyclosporine-A (CsA) on nitric oxide (NO) content and parameters of cardiac function of the diabetic myocardium injured by ischemia/reperfusion. METHODS Diabetes was induced by single injection of streptozotocin (50 mg/kg; intraperitoneally [ip]) in Wistar rats (250-320 g) and the diabetic period was 8 weeks. The hearts (n = 96) were removed quickly, mounted on Langendorff apparatus, and then subjected to 30-minute regional ischemia followed by 45-minute reperfusion. Ischemic postconditioning was induced by 3 cycles of 30-second reperfusion/ischemia at the onset of reperfusion. Myocardial function was measured throughout the experiment, and infarct size (IS) was identified by triphenyltetrazolium chloride (TTC) staining. Total amounts of NO metabolites were determined using Griess method and enzyme-linked immunosorbent assay (ELISA) reader. RESULTS Administration of either IPostC or CsA alone in nondiabetic animals significantly improved myocardial function and reduced the ISs (28% ± 1.9% or 23% ± 2.0% vs 41% ± 2.9% of the risk zone [RZ], respectively; P < .01), but they had no effect on diabetic hearts (35% ± 1.8% or 32% ± 2.1% vs 39% ± 3.1%, respectively). In addition, myocardial NO level was significantly increased by IPostC only in nondiabetic animals (P < .01). However, after administration of CsA (5 minutes before and 10 minutes after the onset of reperfusion) in postconditioned animals, the cardioprotective and NO-enhancing effects of IPostC were restored in diabetic rats (IS: 21% ± 1.1% vs 39% ± 3.1%), similar to those in nondiabetic controls (19% ± 1.3% vs 41% ± 2.9%; P < .01). CONCLUSION The present study indicated that IPostC or CsA failed to affect NO levels and failed to protect the diabetic myocardium against ischemia/reperfusion injury. Moreover, concomitant administration of CsA and IPostC at reperfusion can increase NO content and protect the diabetic myocardium.
Collapse
Affiliation(s)
- Reza Badalzadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | | | | | |
Collapse
|