1
|
Kolb A, Corridon P, Ullah M, Pfaffenberger ZJ, Xu WM, Winfree S, Sandoval RH, Hato T, Witzmann FA, Mohallem R, Franco J, Aryal UK, Atkinson SJ, Basile DP, Bacallao RL. Sulfotransferase 1C2 Increases Mitochondrial Respiration by Converting Mitochondrial Membrane Cholesterol to Cholesterol Sulfate. Biochemistry 2024; 63:2310-2322. [PMID: 39194960 PMCID: PMC11411706 DOI: 10.1021/acs.biochem.3c00344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
HYPOTHESIS In this communication, we test the hypothesis that sulfotransferase 1C2 (SULT1C2, UniProt accession no. Q9WUW8) can modulate mitochondrial respiration by increasing state-III respiration. METHODS AND RESULTS Using freshly isolated mitochondria, the addition of SULT1C2 and 3-phosphoadenosine 5 phosphosulfate (PAPS) results in an increased maximal respiratory capacity in response to the addition of succinate, ADP, and rotenone. Lipidomics and thin-layer chromatography of mitochondria treated with SULT1C2 and PAPS showed an increase in the level of cholesterol sulfate. Notably, adding cholesterol sulfate at nanomolar concentration to freshly isolated mitochondria also increases maximal respiratory capacity. In vivo studies utilizing gene delivery of SULT1C2 expression plasmids to kidneys result in increased mitochondrial membrane potential and confer resistance to ischemia/reperfusion injury. Mitochondria isolated from gene-transduced kidneys have elevated state-III respiration as compared with controls, thereby recapitulating results obtained with mitochondrial fractions treated with SULT1C2 and PAPS. CONCLUSION SULT1C2 increases mitochondrial respiratory capacity by modifying cholesterol, resulting in increased membrane potential and maximal respiratory capacity. This finding uncovers a unique role of SULT1C2 in cellular physiology and extends the role of sulfotransferases in modulating cellular metabolism.
Collapse
Affiliation(s)
- Alexander
J. Kolb
- Department
of Biology, School of Science, Indiana University-Indianapolis, Indianapolis, Indiana 46202, United States
| | - Peter Corridon
- Khalifa
University of Science and Technology, P.O. Box 127788, Abu Dhabi, United Arab Emirates
| | - Mahbub Ullah
- Department
of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | | | - Wei Min Xu
- Division
of Nephrology, Indiana Center for Biological Microscopy, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Seth Winfree
- Department
of Pathology and Microbiology, University
of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Ruben H. Sandoval
- Division
of Nephrology, Indiana Center for Biological Microscopy, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Takeshi Hato
- Division
of Nephrology, Indiana Center for Biological Microscopy, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Frank A. Witzmann
- Department
of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Rodrigo Mohallem
- Department
of Comparative Pathobiology, College of Veterinary Medicine, Bindley
Bioscience Center, Purdue University, West Lafayette, Indiana 47907, United States
| | | | - Uma K. Aryal
- Department
of Comparative Pathobiology, College of Veterinary Medicine, Bindley
Bioscience Center, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue
Proteomics Facility, Bindley Biosciences Center, Purdue University, West Lafayette, Indiana 47907, United States
| | - Simon J. Atkinson
- Department
of Neuroscience, Physiology and Behavior, University of California, Davis, California 95616, United States
| | - David P. Basile
- Department
of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Robert L. Bacallao
- Division
of Nephrology, Indiana Center for Biological Microscopy, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
- Division
of Nephrology, Richard Roudebush
VA Medical Center, Indianapolis, Indiana 46202, United States
| |
Collapse
|
2
|
Wang Q, Xiao J, Wei S, Yang X, Li J, Zuo Y, Hu Z. Remote liver ischemic preconditioning protects against renal ischemia/reperfusion injury via phosphorylation of extracellular signal-regulated kinases 1 and 2 in mice. PLoS One 2024; 19:e0308977. [PMID: 39159207 PMCID: PMC11332924 DOI: 10.1371/journal.pone.0308977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 08/02/2024] [Indexed: 08/21/2024] Open
Abstract
Perioperative acute kidney injury (AKI), which is mainly mediated by renal ischemia‒reperfusion (I/R) injury, is commonly observed in clinical practice. However, effective measures for preventing and treating this perioperative complication are still lacking in the clinic. Thus, we designed this study to examine whether remote liver ischemic preconditioning (RLIPC) has a protective effect on damage caused by renal I/R injury. In a rodent model, 30 mice were divided into five groups to assess the effects of RLIPC and ERK1/2 inhibition on AKI. The groups included the sham-operated (sham), kidney ischemia and reperfusion (CON), remote liver ischemic preconditioning (RLIPC), CON with the ERK1/2 inhibitor U0126 (CON+U0126), and RLIPC with U0126 (RLIPC+U0126). RLIPC consisted of 4 liver ischemia cycles before renal ischemia. Renal function and injury were assessed through biochemical assays, histology, cell apoptosis and protein phosphorylation analysis. RLIPC significantly mitigated renal dysfunction, tissue damage, inflammation, and apoptosis caused by I/R, which was associated with ERK1/2 phosphorylation. Furthermore, ERK1/2 inhibition with U0126 negated the protective effects of RLIPC and exacerbated renal injury. To summarize, we demonstrated that RLIPC has a strong renoprotective effect on kidneys post I/R injury and that this effect may be mediated by phosphorylation of ERK1/2.
Collapse
Affiliation(s)
- Qifeng Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junshen Xiao
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shichao Wei
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xi Yang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiaxue Li
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yunxia Zuo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhaoyang Hu
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Yan Y, Zhao C, Niu J, Yan P, Li J, Wang D, Li G. Rationale and Design of the IMPROVE Trial: A Multicenter, Randomized, Controlled, Open-label, Blinded-endpoint Trial Assessing the Efficacy of Remote Ischemic Conditioning in Patients Undergoing Off-Pump Coronary Artery Bypass Grafting. Adv Ther 2024; 41:3003-3012. [PMID: 38616242 DOI: 10.1007/s12325-024-02836-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 03/07/2024] [Indexed: 04/16/2024]
Abstract
INTRODUCTION Despite the appearance of off-pump coronary artery bypass grafting (CABG), ischemia-reperfusion injury (IRI) in the perioperative period still arouses concerns of clinicians. Remote ischemic conditioning (RIC) is the process of repeated ischemia and reperfusion in the peripheral vessels, which is proven to reduce IRI in vital organs. However, the effect of RIC in patients undergoing off-pump CABG is still unclear. METHODS This IMPROVE trial is a national, multicenter, randomized, controlled, open-label, blinded-endpoint clinical trial designed to assess whether RIC intervention can improve short-term prognosis of patients undergoing off-pump CABG. It plans to enroll 648 patients who will be randomly assigned into a RIC group or control group. Patients in the RIC group will receive four cycles of 5 min of pressurization (about 200 mmHg) and 5 min of rest in the 3 days before and 7 days after the surgery. PLANNED OUTCOMES The primary outcome is the occurrence of major adverse cardiovascular and cerebrovascular events (MACCE) within the 3-month follow-up. MACCE is defined as all-cause death, myocardial infarction, stroke, and coronary revascularization surgery. CLINICAL TRIAL REGISTRATION NCT06141525 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Yang Yan
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Changying Zhao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jialan Niu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Pengyun Yan
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jing Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Cardiovascular Surgery, People's Hospital of Qinghai Province, Xining, China
| | - Duolao Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, UK
- Department of Neurology, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Guoliang Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
4
|
Barrère-Lemaire S, Vincent A, Jorgensen C, Piot C, Nargeot J, Djouad F. Mesenchymal stromal cells for improvement of cardiac function following acute myocardial infarction: a matter of timing. Physiol Rev 2024; 104:659-725. [PMID: 37589393 DOI: 10.1152/physrev.00009.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/05/2023] [Accepted: 08/16/2023] [Indexed: 08/18/2023] Open
Abstract
Acute myocardial infarction (AMI) is the leading cause of cardiovascular death and remains the most common cause of heart failure. Reopening of the occluded artery, i.e., reperfusion, is the only way to save the myocardium. However, the expected benefits of reducing infarct size are disappointing due to the reperfusion paradox, which also induces specific cell death. These ischemia-reperfusion (I/R) lesions can account for up to 50% of final infarct size, a major determinant for both mortality and the risk of heart failure (morbidity). In this review, we provide a detailed description of the cell death and inflammation mechanisms as features of I/R injury and cardioprotective strategies such as ischemic postconditioning as well as their underlying mechanisms. Due to their biological properties, the use of mesenchymal stromal/stem cells (MSCs) has been considered a potential therapeutic approach in AMI. Despite promising results and evidence of safety in preclinical studies using MSCs, the effects reported in clinical trials are not conclusive and even inconsistent. These discrepancies were attributed to many parameters such as donor age, in vitro culture, and storage time as well as injection time window after AMI, which alter MSC therapeutic properties. In the context of AMI, future directions will be to generate MSCs with enhanced properties to limit cell death in myocardial tissue and thereby reduce infarct size and improve the healing phase to increase postinfarct myocardial performance.
Collapse
Affiliation(s)
- Stéphanie Barrère-Lemaire
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Anne Vincent
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Christian Jorgensen
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - Christophe Piot
- Département de Cardiologie Interventionnelle, Clinique du Millénaire, Montpellier, France
| | - Joël Nargeot
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Farida Djouad
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| |
Collapse
|
5
|
Liang H, Ye R, Zhang X, Ye H, Ouyang W, Cai S, Wei L. Autonomic function may mediate the neuroprotection of remote ischemic postconditioning in stroke: A randomized controlled trial. J Stroke Cerebrovasc Dis 2023; 32:107198. [PMID: 37329785 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/18/2023] [Accepted: 05/20/2023] [Indexed: 06/19/2023] Open
Abstract
OBJECTIVES To evaluate the effect of remote ischemic postconditioning (RIPostC) on the prognosis of acute ischemic stroke(AIS) patients and investigate the mediating role of autonomic function in the neuroprotection of RIPostC. MATERIALS AND METHODS 132 AIS patients were randomized into two groups. Patients received four cycles of 5-min inflation to a pressure of 200 mmHg(i.e., RIPostC) or patients' diastolic BP(i.e., shame), followed by 5 min of deflation on healthy upper limbs once a day for 30 days. The main outcome was neurological outcome including the National Institutes of Health Stroke Scale (NIHSS), modified Rankin Scale (mRS), and Barthel index(BI). The second outcome measure was autonomic function measured by heart rate variability(HRV). RESULTS Compared with the baseline, the post-intervention NIHSS score was significantly reduced in both groups (P<0.001). NIHSS score was significantly lower in the control group than intervention group at day 7.[RIPostC:3(1,5) versus shame:2(1,4); P=0.030]. mRS scored lower in the intervention group compared with the control group at day 90 follow-up(RIPostC:0.5±2.0 versus shame:1.0±2.0;P=0.016). The goodness-of-fit test revealed a significant difference between the generalized estimating equation model of mRS and BI scores of uncontrolled-HRV and controlled-HRV(P<0.05, both). The results of bootstrap revealed a complete mediation effect of HRV between group on mRS[indirect effect: -0.267 (LLCI = -0.549, ULCI = -0.048), the direct effect: -0.443 (LLCI = -0.831, ULCI = 0.118)]. CONCLUSION This is the first human-based study providing evidence for a mediation role of autonomic function between RIpostC and prognosis in AIS patients. It indicated that RIPostC could improve the neurological outcome of AIS patients. Autonomic function may play a mediating role in this association. TRIAL REGISTRATION The clinical trials registration number for this study is NCT02777099 (ClinicalTrials.gov Identifier).
Collapse
Affiliation(s)
- Hao Liang
- Department of Neurology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Richun Ye
- Department of Neurology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaopei Zhang
- Department of Neurology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huanwen Ye
- Department of Cardiac Function, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wenwei Ouyang
- Key Unit of Methodology in Clinical Research, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shuang Cai
- Tongde Hospital of Zhejiang Province, Zhejiang, China
| | - Lin Wei
- Department of Neurology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
6
|
Zhu Y, Wang S, Chen X. Extracellular Vesicles and Ischemic Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1418:57-68. [PMID: 37603272 DOI: 10.1007/978-981-99-1443-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Characterized by coronary artery obstruction or stenosis, ischemic cardiovascular diseases as advanced stages of coronary heart diseases commonly lead to left ventricular aneurysm, ventricular septal defect, and mitral insufficiency. Extracellular vesicles (EVs) secreted by diverse cells in the body exert roles in cell-cell interactions and intrinsic cellular regulations. With a lipid double-layer membrane and biological components such as DNA, protein, mRNA, microRNAs (miRNA), and siRNA inside, the EVs function as paracrine signaling for the pathophysiology of ischemic cardiovascular diseases and maintenance of the cardiac homeostasis. Unlike stem cell transplantation with the potential tumorigenicity and immunogenicity, the EV-based therapeutic strategy is proposed to satisfy the demand for cardiac repair and regeneration while the circulating EVs detected by a noninvasive approach can act as precious biomarkers. In this chapter, we extensively summarize the cardioprotective functions of native EVs and bioengineered EVs released from stem cells, cardiomyocytes, cardiac progenitor cells (CPCs), endothelial cells, fibroblast, smooth muscle cells, and immune cells. In addition, the potential of EVs as robust molecule biomarkers is discussed for clinical diagnosis of ischemic cardiovascular disease, attributed to the same pathology of EVs as that of their origin. Finally, we highlight EV-based therapy as a biocompatible alternative to direct cell-based therapy for ischemic cardiovascular diseases.
Collapse
Affiliation(s)
- Yujiao Zhu
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Siqi Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Xuerui Chen
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China.
| |
Collapse
|
7
|
Wu YK, Harel NY, Wecht JM, Bloom OE. Effects of Remote Ischemic Conditioning on Hand Engagement in individuals with Spinal cord Injury (RICHES): protocol for a pilot crossover study. F1000Res 2022; 10:464. [PMID: 35342620 PMCID: PMC8924555 DOI: 10.12688/f1000research.52670.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/06/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Most spinal cord injuries (SCI) are not full transections, indicating that residual nerve circuits are retained. Rehabilitation interventions have been shown to beneficially reorganize motor pathways in the brain, corticospinal tract, and at the spinal level. However, rehabilitation training require a large number of repetitions, and intervention effects may be absent or show transient retention. Therefore, the need remains for an effective approach to synergistically improve the amount and duration of neuroplasticity in combination with other interventions. Remote ischemic conditioning (RIC) demonstrates several potential advantages as a candidate for such an approach. Therefore, we propose a protocol to investigate RIC coupled with physical training to promote neuroplasticity in hand muscles. Methods: This will be a prospective randomized-order crossover trial to be performed in eight able-bodied participants and eight participants with chronic cervical SCI. Patients will participate in two experimental sessions consisting of either active or sham RIC preceding a bout of pinch movement exercise. Serial evaluations will be conducted at baseline, after RIC, immediately after pinch exercise, and follow up 15-minutes later. The primary outcome is the change in corticospinal excitability (primarily measured by the motor evoked potential of abductor pollicis brevis muscle). Secondary outcomes will include maximal volitional pinch force, and inflammatory biomarkers. To ensure safety, we will monitor tolerability and hemodynamic responses during RIC. Discussion: This protocol will be the first to test RIC in people with cervical SCI and to investigate whether RIC alters corticospinal excitability. By sharing the details of our protocol, we hope other interested researchers will seek to investigate similar approaches – depending on overlap with the current study and mutual sharing of participant-level data, this could increase the sample size, power, and generalizability of the analysis and results. Trial registration: ClinicalTrial.gov, ID: NCT03851302; Date of registration: February 22, 2019
Collapse
Affiliation(s)
- Yu-Kuang Wu
- Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York, NY, 10003, USA
- Bronx Veterans Medical Research Foundation, Bronx, NY, 10468, USA
| | - Noam Y. Harel
- Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York, NY, 10003, USA
- Bronx Veterans Medical Research Foundation, Bronx, NY, 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10003, USA
| | - Jill M. Wecht
- Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York, NY, 10003, USA
- Bronx Veterans Medical Research Foundation, Bronx, NY, 10468, USA
| | - Ona E. Bloom
- Bronx Veterans Medical Research Foundation, Bronx, NY, 10468, USA
- The Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA
- The Zucker School of Medicine at Hofstra Northwell, Hempstead, NY, 11549, USA
| |
Collapse
|
8
|
Wu J, Yu C, Zeng X, Sun C. The hepatoprotective effect from ischemia-reperfusion injury of remote ischemic preconditioning in the liver related surgery: a meta-analysis. ANZ J Surg 2021; 92:1332-1337. [PMID: 34854193 DOI: 10.1111/ans.17236] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND This study aimed to assess the hepatoprotective effect of remote ischemic preconditioning (RIPC) in the liver related surgery. METHODS Published articles in PubMed, Embase and Cochrane clinical trial databases were searched from the inception to May 2021. Randomized control trials (RCTs) comparing the RIPC with control or other conditionings were included for analysis. The postoperative liver synthetic function was used as the primary outcome. RESULTS A total of six RCTs were included the present meta-analysis. There were 216 patients underwent RIPC and 212 patients in the control group. The RIPC group had a significantly lower level of postoperative alanine transaminase and aspartate transaminase (p<0.001). The postoperative bilirubin level was also significant lower in the RIPC group than the control group (MD = -9.0, 95%CI, -13.94 to -4.03; p<0.001). ICG clearance was reduced in controls versus RIPC (p<0.001). There was no significant difference between the RIPC and control group in terms of the complication rate. CONCLUSION The RIPC was evaluated to have a strong hepatoprotective effect from ischemia-reperfusion injury in the liver related surgery.
Collapse
Affiliation(s)
- Jinli Wu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Chao Yu
- Department of General Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xianggang Zeng
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Chengyi Sun
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Soochow, China
| |
Collapse
|
9
|
Mi L, Zhang N, Wan J, Cheng M, Liao J, Zheng X. Remote ischemic post‑conditioning alleviates ischemia/reperfusion‑induced intestinal injury via the ERK signaling pathway‑mediated RAGE/HMGB axis. Mol Med Rep 2021; 24:773. [PMID: 34490475 PMCID: PMC8441982 DOI: 10.3892/mmr.2021.12413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022] Open
Abstract
Intestinal ischemia reperfusion (I/R) injury is a tissue and organ injury that frequently occurs during surgery and significantly contributes to the pathological processes of severe infection, injury, shock, cardiopulmonary insufficiency and other diseases. However, the mechanism of intestinal I/R injury remains to be elucidated. A mouse model of intestinal I/R injury was successfully established and the model mice were treated with remote ischemic post‑conditioning (RIPOC) and/or an ERK inhibitor (CC‑90003), respectively. Histopathological changes of the intestinal mucosa were determined by hematoxylin and eosin staining. In addition, the levels of high‑mobility group box 1 (HMGB1) and receptor for advanced glycation end products (RAGE) expression were confirmed by reverse transcription‑quantitative polymerase chain reaction, western blotting and immunohistochemistry assays. The levels of antioxidants, oxidative stress markers (8‑OHdG) and interleukin 1 family members were evaluated by ELISA assays and the levels of NF‑κB pathway proteins were analyzed by western blotting. The data demonstrated that RIPOC could attenuate the histopathological features of intestinal mucosa in the intestinal I/R‑injury mouse models via the ERK pathway. It was also revealed that HMGB1 and RAGE expression in the mouse models could be markedly reduced by RIPOC (P<0.05) and that these reductions were associated with inhibition of the ERK pathway. Furthermore, it was demonstrated that RIPOC produced significant antioxidant and anti‑inflammatory effects following an intestinal I/R injury and that these effects were mediated via the ERK pathway (P<0.05). In addition, RIPOC was demonstrated to suppress the NF‑κB (p65)/NLR family pyrin domain containing 3 (NLRP3) inflammatory pathways in the intestinal I/R injury mouse models via the ERK pathway. The findings of the present study demonstrated that RIPOC helped to protect mice with an intestinal I/R injury by downregulating the ERK pathway.
Collapse
Affiliation(s)
- Lei Mi
- Department of Gastrointestinal Surgery, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Nan Zhang
- Department of Gastrointestinal Surgery, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Jiyun Wan
- Department of Gastrointestinal Surgery, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Ming Cheng
- Department of Gastrointestinal Surgery, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Jianping Liao
- Department of Gastrointestinal Surgery, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Xiao Zheng
- Department of Gastrointestinal Surgery, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| |
Collapse
|
10
|
Pansani AP, Ghazale PP, Dos Santos EG, Dos Santos Borges K, Gomes KP, Lacerda IS, Castro CH, Mendes EP, Dos Santos FCA, Biancardi MF, Nejm MB, Dogini DB, Rabelo LA, Nunes-Souza V, Scorza FA, Colugnati DB. The number and periodicity of seizures induce cardiac remodeling and changes in micro-RNA expression in rats submitted to electric amygdala kindling model of epilepsy. Epilepsy Behav 2021; 116:107784. [PMID: 33548915 DOI: 10.1016/j.yebeh.2021.107784] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/03/2021] [Accepted: 01/03/2021] [Indexed: 12/20/2022]
Abstract
Generalized tonic-clonic seizures (GTCS) are the main risk factor for sudden unexpected death in epilepsy (SUDEP). Also, among the several mechanisms underlying SUDEP there is the cardiac dysfunction. So, we aimed to evaluate the impact of the number of seizures on heart function and morphology in rats with epilepsy. Rats were randomized into three groups: Sham (without epilepsy), 5 S, and 10 S groups, referred as rats with epilepsy with a total of 5 or 10 GTCS, respectively. Epilepsy was induced by electrical amygdala kindling. The ventricular function was analyzed by the Langendorff technique and challenged by ischemia/reperfusion protocol. Cardiac fibrosis and hypertrophy were analyzed by histology. We also analyzed cardiac metalloproteinases (MMP2 and MMP9), ERK 1/2 and phosphorylated ERK1/2 (P-ERK) by western blot; microRNA-21 and -320 by RT-PCR; and oxidative stress (TBARS, catalase activity and nitrite) by biochemical analysis. Only the 5S group presented decreased values of ventricular function at before ischemia/reperfusion (baseline): intraventricular systolic pressure, developed intraventricular pressure, positive and negative dP/dt. During ischemia/reperfusion protocol, the variation of the ventricular function did not differ among groups. Both 5S and 10S groups had increased cardiomyocyte hypertrophy and fibrosis compared to Sham, but in the 5S group, these alterations were higher than in the 10S group. The 5S group increased in microRNA-21 and decreased in microRNA-320 expression compared to Sham and the 10S group. The 10S group increased in MMP9 and decreased in P-ERK/ERK expression, and increased in nitrite content compared to both Sham and the 5S group. Therefore, seizures impair cardiac function and morphology, probably through microRNA modulation. The continuation of seizures seems to exert a preconditioning-like stimulus that fails to compensate the cardiac tissue alteration.
Collapse
Affiliation(s)
- Aline Priscila Pansani
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil.
| | - Poliana Peres Ghazale
- Department of Neurology and Neurosurgery, Federal University of São Paulo, São Paulo, Brazil
| | - Emilly Gomes Dos Santos
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Kiscilla Dos Santos Borges
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Karina Pereira Gomes
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Ismaley Santos Lacerda
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Carlos Henrique Castro
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Elizabeth Pereira Mendes
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | | | | | - Mariana Bocca Nejm
- Department of Neurology and Neurosurgery, Federal University of São Paulo, São Paulo, Brazil
| | - Danyella Barbosa Dogini
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Luiza Antas Rabelo
- Department of Physiology, Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, Alagoas, Brazil
| | - Valéria Nunes-Souza
- Department of Physiological and Pharmacology Sciences, Institute of Biological Sciences, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Fulvio Alexandre Scorza
- Department of Neurology and Neurosurgery, Federal University of São Paulo, São Paulo, Brazil
| | - Diego Basile Colugnati
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| |
Collapse
|
11
|
Zhong SJ, Cui MM, Gao YT, Cao XY, Chen B, Wen XR. MicroRNA-144 promotes remote limb ischemic preconditioning-mediated neuroprotection against ischemic stroke via PTEN/Akt pathway. Acta Neurol Belg 2021; 121:95-106. [PMID: 32960423 DOI: 10.1007/s13760-020-01500-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 09/10/2020] [Indexed: 12/18/2022]
Abstract
Ischemic stroke is a refractory disease generally caused by cerebral ischemic injury. Remote ischemic preconditioning (RIPC) caused by transient ischemia and reperfusion of the femoral artery exerts a protective effect on ischemic stroke-induced brain injury. This study was designed to investigate the potential molecular mechanism of RIPC-mediated neuroprotection, namely, the biological effects of microRNA-144 on RIPC in mice with ischemic stroke and its effects on PTEN and Akt signaling pathways. Healthy adult C57BL6 mice were selected for the establishment of middle cerebral artery occlusion (MCAO). One hour before the start, remote ischemic preconditioning of limbs was performed in mice. Brain edema and infarct volume were measured. The expressions of microRNA-144, PTEN, and Akt were measured. The results showed that, compared with MCAO group, the RIPC group protected mice from cerebral ischemia-reperfusion injury, systemic accumulation of inflammatory cytokines, and accelerated apoptosis of parenchymal cells. In RIPC group, PTEN expression decreased, and mir-144 and Akt expression increased. The level of phosphorylated PTEN in the transfected microRNA-144 inhibitor group increased and the level of phosphorylated Akt reduced significantly. In conclusion, our results suggest that microRNA-144 may play a protective role in remote ischemic pretreatment by downregulating PTEN and upregulating Akt, suggesting that microRNA-144 via PTEN/Akt pathway may be of therapeutic significance in ischemic stroke.
Collapse
Affiliation(s)
- Si-Jin Zhong
- Department of Clinical, Xuzhou Medical University, Xuzhou, 221004, China
| | - Miao-Miao Cui
- Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yu-Ting Gao
- Medical Technology School, Xuzhou Medical University, Xuzhou, 221004, China
| | - Xue-Yan Cao
- Department of Clinical, Xuzhou Medical University, Xuzhou, 221004, China
| | - Bin Chen
- Department of Rehabilitation and National Clinical Research Base of Traditional Chinese Medicine, The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, 350004, China.
| | - Xian-Ru Wen
- Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China.
| |
Collapse
|
12
|
Wei L, Liang H, Mo M, Liu Z, Ye R, Ye H, Ouyang W, Yu W, Zhao W, Zhang X. The effect of remote ischemic postconditioning on autonomic function in patients with acute ischemic stroke: A Randomized Controlled Trail. Complement Ther Med 2020; 54:102541. [PMID: 33183660 DOI: 10.1016/j.ctim.2020.102541] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/22/2020] [Accepted: 08/13/2020] [Indexed: 01/29/2023] Open
Abstract
OBJECTIVE The evidence for the effect of remote ischemic postconditioning(RIpostC) on autonomic function in patients with acute ischemic stroke(AIS) is lacking and the neural mechanism underlying the protection of RIpostC remains speculative. This trial was aimed to evaluated the efficiency of RIpostC on autonomic function in AIS patients. DESIGN One hundred and six AIS patients were included in this prospective, randomized, placebo-controlled trial. Patients in intervention group (n = 57) received 4 cycles of alternating inflation (cuff inflation to 200 mmHg) and deflation for 5 min on healthy upper arm once a day for 30 days. The control group underwent a sham inflation and deflation cycles. Autonomic function was evaluated by heart rate variability (HRV). RESULTS All HRV parameters except for the ratio of low frequency to high frequency (P = 0.101) increased significantly with time (P < 0.001) in the two groups. The value of standard deviation of all normal R-R intervals(SDNN) and high frequency at day7 and day30 and the value of the percent of difference between adjacent normal R-R intervals (pNN50) at day 30 in RIpostC group was significantly higher than that of the sham-RIpostC group(P < 0.05). A significant time-by-group interaction was observed in SDNN、pNN50、and high frequency over time between two groups (P < 0.05). CONCLUSIONS 30-day RIpostC could improve autonomic function in AIS patients through the enhancement of the total autonomic nerve activity and vagus nerve activity. The mechanism of RIpostC mediating autonomic function needs to be further investigated.
Collapse
Affiliation(s)
- Lin Wei
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Dade Road 111, Yuexiu District, Guangzhou 510120, Guangdong, China
| | - Hao Liang
- School of Nursing, Guangzhou University of Chinese Medicine, Airport Road 12, Baiyun District, Guangzhou 510405, Guangdong, China
| | - Miaomiao Mo
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Dade Road 111, Yuexiu District, Guangzhou 510120, Guangdong, China
| | - Zhuyun Liu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Dade Road 111, Yuexiu District, Guangzhou 510120, Guangdong, China
| | - Richun Ye
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Dade Road 111, Yuexiu District, Guangzhou 510120, Guangdong, China
| | - Huanwen Ye
- Department of Cardiac Function, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Dade Road 111, Yuexiu District, Guangzhou 510120, Guangdong, China
| | - Wenwei Ouyang
- Key Unit of Methodology in Clinical Research, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Dade Road 111, Yuexiu District, Guangzhou 510120, Guangdong, China
| | - Wenqi Yu
- Geriatrics dept(neurology), The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Inner Ring West Road 55, Panyu District, Guangzhou 510006, Guangdong, China
| | - Wenbo Zhao
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-Sen University, Tianhe Road 600, Tianhe District, Guangzhou 510632, Guangdong, China.
| | - Xiaopei Zhang
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Dade Road 111, Yuexiu District, Guangzhou 510120, Guangdong, China.
| |
Collapse
|
13
|
Effect of Remote Ischemic Preconditioning Conducted in Living Liver Donors on Postoperative Liver Function in Donors and Recipients Following Liver Transplantation: A Randomized Clinical Trial. Ann Surg 2020; 271:646-653. [PMID: 31356262 DOI: 10.1097/sla.0000000000003498] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE This study aimed to assess the effects of remote ischemic preconditioning (RIPC) on liver function in donors and recipients after living donor liver transplantation (LDLT). BACKGROUND Ischemia reperfusion injury (IRI) is known to be associated with graft dysfunction after liver transplantation. RIPC is used to lessen the harmful effects of IRI. METHODS A total of 148 donors were randomly assigned to RIPC (n = 75) and control (n = 73) groups. RIPC involves 3 cycles of 5-minute inflation of a blood pressure cuff to 200 mm Hg to the upper arm, followed by 5-minute reperfusion with cuff deflation. The primary aim was to assess postoperative liver function in donors and recipients and the incidence of early allograft dysfunction and graft failure in recipients. RESULTS RIPC was not associated with any differences in postoperative aspartate aminotransferase (AST) and alanine aminotransferase levels after living donor hepatectomy, and it did not decrease the incidence of delayed graft hepatic function (6.7% vs 0.0%, P = 0.074) in donors. AST level on postoperative day 1 [217.0 (158.0, 288.0) vs 259.5 (182.0, 340.0), P = 0.033] and maximal AST level within 7 postoperative days [244.0 (167.0, 334.0) vs 296.0 (206.0, 395.5), P = 0.029) were significantly lower in recipients who received a preconditioned graft. No differences were found in the incidence of early allograft dysfunction (4.1% vs 5.6%, P = 0.955) or graft failure (1.4% vs 5.6%, P = 0.346) among recipients. CONCLUSIONS RIPC did not improve liver function in living donor hepatectomy. However, RIPC performed in liver donors may be beneficial for postoperative liver function in recipients after living donor liver transplantation.
Collapse
|
14
|
Zhou D, Ding J, Ya J, Pan L, Wang Y, Ji X, Meng R. Remote ischemic conditioning: a promising therapeutic intervention for multi-organ protection. Aging (Albany NY) 2019; 10:1825-1855. [PMID: 30115811 PMCID: PMC6128414 DOI: 10.18632/aging.101527] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 08/10/2018] [Indexed: 12/21/2022]
Abstract
Despite decades of formidable exploration, multi-organ ischemia-reperfusion injury (IRI) encountered, particularly amongst elderly patients with clinical scenarios, such as age-related arteriosclerotic vascular disease, heart surgery and organ transplantation, is still an unsettled conundrum that besets clinicians. Remote ischemic conditioning (RIC), delivered via transient, repetitive noninvasive IR interventions to distant organs or tissues, is regarded as an innovative approach against IRI. Based on the available evidence, RIC holds the potential of affording protection to multiple organs or tissues, which include not only the heart and brain, but also others that are likely susceptible to IRI, such as the kidney, lung, liver and skin. Neuronal and humoral signaling pathways appear to play requisite roles in the mechanisms of RIC-related beneficial effects, and these pathways also display inseparable interactions with each other. So far, several hurdles lying ahead of clinical translation that remain to be settled, such as establishment of biomarkers, modification of RIC regimen, and deep understanding of underlying minutiae through which RIC exerts its powerful function. As this approach has garnered an increasing interest, herein, we aim to encapsulate an overview of the basic concept and postulated protective mechanisms of RIC, highlight the main findings from proof-of-concept clinical studies in various clinical scenarios, and also to discuss potential obstacles that remain to be conquered. More well designed and comprehensive experimental work or clinical trials are warranted in future research to confirm whether RIC could be utilized as a non-invasive, inexpensive and efficient adjunct therapeutic intervention method for multi-organ protection.
Collapse
Affiliation(s)
- Da Zhou
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Jiayue Ding
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Jingyuan Ya
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Liqun Pan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Yuan Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Ran Meng
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Geriatric Disorders, Beijing, China
| |
Collapse
|
15
|
Jiang J, Hoagland D, Palatinus JA, He H, Iyyathurai J, Jourdan LJ, Bultynck G, Wang Z, Zhang Z, Schey K, Poelzing S, McGowan FX, Gourdie RG. Interaction of α Carboxyl Terminus 1 Peptide With the Connexin 43 Carboxyl Terminus Preserves Left Ventricular Function After Ischemia-Reperfusion Injury. J Am Heart Assoc 2019; 8:e012385. [PMID: 31422747 PMCID: PMC6759879 DOI: 10.1161/jaha.119.012385] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Background α Carboxyl terminus 1 (αCT1) is a 25–amino acid therapeutic peptide incorporating the zonula occludens‐1 (ZO‐1)–binding domain of connexin 43 (Cx43) that is currently in phase 3 clinical testing on chronic wounds. In mice, we reported that αCT1 reduced arrhythmias after cardiac injury, accompanied by increases in protein kinase Cε phosphorylation of Cx43 at serine 368. Herein, we characterize detailed molecular mode of action of αCT1 in mitigating cardiac ischemia‐reperfusion injury. Methods and Results To study αCT1‐mediated increases in phosphorylation of Cx43 at serine 368, we undertook mass spectrometry of protein kinase Cε phosphorylation assay reactants. This indicated potential interaction between negatively charged residues in the αCT1 Asp‐Asp‐Leu‐Glu‐Iso sequence and lysines (Lys345, Lys346) in an α‐helical sequence (helix 2) within the Cx43‐CT. In silico modeling provided further support for this interaction, indicating that αCT1 may interact with both Cx43 and ZO‐1. Using surface plasmon resonance, thermal shift, and phosphorylation assays, we characterized a series of αCT1 variants, identifying peptides that interacted with either ZO‐1–postsynaptic density‐95/disks large/zonula occludens‐1 2 or Cx43‐CT, but with limited or no ability to bind both molecules. Only peptides competent to interact with Cx43‐CT, but not ZO‐1–postsynaptic density‐95/disks large/zonula occludens‐1 2 alone, prompted increased pS368 phosphorylation. Moreover, in an ex vivo mouse model of ischemia‐reperfusion injury, preischemic infusion only with those peptides competent to bind Cx43 preserved ventricular function after ischemia‐reperfusion. Interestingly, a short 9–amino acid variant of αCT1 (αCT11) demonstrated potent cardioprotective effects when infused either before or after ischemic injury. Conclusions Interaction of αCT1 with the Cx43, but not ZO‐1, is correlated with cardioprotection. Pharmacophores targeting Cx43‐CT could provide a translational approach to preserving heart function after ischemic injury.
Collapse
Affiliation(s)
- Jingbo Jiang
- Fralin Biomedical Research Institute at Virginia Tech Carilion Center for Heart and Reparative Medicine Research Virginia Tech Blacksburg VA.,Shenzhen Children's Hospital Shenzhen China.,Department of Pediatric Cardiology Guangdong Cardiovascular Institute Guangdong General Hospital Guangdong Academy of Medical Sciences Guangzhou China
| | - Daniel Hoagland
- Fralin Biomedical Research Institute at Virginia Tech Carilion Center for Heart and Reparative Medicine Research Virginia Tech Blacksburg VA
| | - Joseph A Palatinus
- Cedars-Sinai Heart Smidt Institute Cedars-Sinai Medical Center Los Angeles CA
| | - Huamei He
- Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia and University of Pennsylvania Philadelphia PA
| | - Jegan Iyyathurai
- Department Cellular and Molecular Medicine KU Leuven Laboratory of Molecular and Cellular Signaling Leuven Belgium
| | - L Jane Jourdan
- Fralin Biomedical Research Institute at Virginia Tech Carilion Center for Heart and Reparative Medicine Research Virginia Tech Blacksburg VA
| | - Geert Bultynck
- Department Cellular and Molecular Medicine KU Leuven Laboratory of Molecular and Cellular Signaling Leuven Belgium
| | - Zhen Wang
- Department of Biochemistry Vanderbilt University School of Medicine Nashville TN
| | - Zhiwei Zhang
- Department of Pediatric Cardiology Guangdong Cardiovascular Institute Guangdong General Hospital Guangdong Academy of Medical Sciences Guangzhou China
| | - Kevin Schey
- Department of Biochemistry Vanderbilt University School of Medicine Nashville TN
| | - Steven Poelzing
- Fralin Biomedical Research Institute at Virginia Tech Carilion Center for Heart and Reparative Medicine Research Virginia Tech Blacksburg VA.,Department of Biomedical Engineering and Mechanics Virginia Tech Blacksburg VA
| | - Francis X McGowan
- Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia and University of Pennsylvania Philadelphia PA
| | - Robert G Gourdie
- Fralin Biomedical Research Institute at Virginia Tech Carilion Center for Heart and Reparative Medicine Research Virginia Tech Blacksburg VA.,Department of Biomedical Engineering and Mechanics Virginia Tech Blacksburg VA
| |
Collapse
|
16
|
Tsibulnikov SY, Maslov LN, Gorbunov AS, Voronkov NS, Boshchenko AA, Popov SV, Prokudina ES, Singh N, Downey JM. A Review of Humoral Factors in Remote Preconditioning of the Heart. J Cardiovasc Pharmacol Ther 2019; 24:403-421. [PMID: 31035796 DOI: 10.1177/1074248419841632] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A humoral mechanism of cardioprotection by remote ischemic preconditioning (RIP) has been clearly demonstrated in various models of ischemia-reperfusion including upper and lower extremities, liver, and the mesenteric and renal arteries. A wide range of humoral factors for RIP have been proposed including hydrophobic peptides, opioid peptides, adenosine, prostanoids, endovanilloids, endocannabinoids, calcitonin gene-related peptide, leukotrienes, noradrenaline, adrenomedullin, erythropoietin, apolipoprotein, A-I glucagon-like peptide-1, interleukin 10, stromal cell-derived factor 1, and microRNAs. Virtually, all of the components of ischemic preconditioning's signaling pathway such as nitric oxide synthase, protein kinase C, redox signaling, PI3-kinase/Akt, glycogen synthase kinase β, ERK1/2, mitoKATP channels, Connexin 43, and STAT were all found to play a role. The signaling pattern also depends on which remote vascular bed was subjected to ischemia and on the time between applying the rip and myocardial ischemia occurs. Because there is convincing evidence for many seemingly diverse humoral components in RIP, the most likely explanation is that the overall mechanism is complex like that seen in ischemic preconditioning where multiple components are both in series and in parallel and interact with each other. Inhibition of any single component in the right circumstance may block the resulting protective effect, and selectively activating that component may trigger the protection. Identifying the humoral factors responsible for RIP might be useful in developing drugs that confer RIP's protection in a more comfortable and reliable manner.
Collapse
Affiliation(s)
- Sergey Y Tsibulnikov
- 1 Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Tomsk, Russia
| | - Leonid N Maslov
- 1 Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Tomsk, Russia
| | - Alexander S Gorbunov
- 1 Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Tomsk, Russia
| | - Nikita S Voronkov
- 1 Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Tomsk, Russia
| | - Alla A Boshchenko
- 1 Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Tomsk, Russia
| | - Sergey V Popov
- 1 Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Tomsk, Russia
| | - Ekaterina S Prokudina
- 1 Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Tomsk, Russia
| | - Nirmal Singh
- 2 Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - James M Downey
- 3 Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| |
Collapse
|
17
|
Sipahi M, Gunaydin M, Kesicioglu T, Usta M, Yavuz BT, Tomruk C. A new approach to prevent ischemia/reperfusion injury in a rat model: remote ischemic conditioning. Arch Gynecol Obstet 2019; 299:1691-1699. [DOI: 10.1007/s00404-019-05149-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 04/03/2019] [Indexed: 01/19/2023]
|
18
|
Rise N, Kristiansen J, Hvas AM, Grove EL, Würtz M, Neergaard-Petersen S, Kristensen SD. Effect of remote ischaemic conditioning on platelet aggregation and platelet turnover. J Thromb Thrombolysis 2018; 46:528-533. [PMID: 30168042 DOI: 10.1007/s11239-018-1728-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Remote ischaemic conditioning (RIC) is a new beneficial treatment for patients with ST-elevation myocardial infarction. RIC may inhibit thrombus formation and, therefore, we investigated whether RIC affects platelet aggregation and turnover. 30 healthy male volunteers were subjected to intervention on day 1 (sham intervention, no aspirin), day 2 (RIC, no aspirin), and day 16 (RIC, treated 7 days with aspirin 75 mg/day). RIC was performed as four cycles of 5 min interchangeable inflation and deflation using an automated cuff. Blood samples were collected 5 min before, as well as 5 and 45 min after RIC. Platelet aggregation was measured by Multiplate® using collagen (COLtest), adenosine diphosphate (ADPtest), and arachidonic acid (ASPItest) as agonists. Platelet turnover was evaluated by flow cytometry. Serum thromboxane B2 was determined by ELISA to confirm aspirin compliance. We found no significant change in platelet aggregation at visit 1 (COLtest: p = 0.32; ADPtest: p = 0.24; ASPItest: p = 0.07), visit 2, except for ADP-induced platelet aggregation evaluated 5 min after RIC (COLtest: p = 0.39; ADPtest: p = 0.02; ASPItest: p = 0.39), or visit 3 (COLtest: p = 0.48; ADPtest: p = 0.61; ASPItest: p = 0.90). Platelet turnover was not influenced by RIC, neither on nor off aspirin (all p-values > 0.07). (1) RIC did not affect platelet aggregation in healthy young men. (2) RIC did not affect platelet turnover in healthy young men. (3) Aspirin did not influence the effect of RIC on platelet aggregation and turnover. (4) Future studies exploring the effect of RIC on platelet aggregation and turnover in patients with ischaemic heart disease are warranted.
Collapse
Affiliation(s)
- Nina Rise
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus, Denmark
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus, Denmark
| | - Jacobina Kristiansen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus, Denmark
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus, Denmark
| | - Anne-Mette Hvas
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200, Aarhus, Denmark
| | - Erik L Grove
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200, Aarhus, Denmark
| | - Morten Würtz
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus, Denmark
| | - Søs Neergaard-Petersen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus, Denmark
| | - Steen Dalby Kristensen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus, Denmark.
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200, Aarhus, Denmark.
| |
Collapse
|
19
|
The Possible Pathophysiological Outcomes and Mechanisms of Tourniquet-Induced Ischemia-Reperfusion Injury during Total Knee Arthroplasty. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8087598. [PMID: 30524661 PMCID: PMC6247434 DOI: 10.1155/2018/8087598] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/04/2018] [Indexed: 12/22/2022]
Abstract
Ischemia and reperfusion (I/R) injury induced by tourniquet (TQ) application leads to the release of both oxygen free radicals and inflammatory cytokines. The skeletal muscle I/R may contribute to local skeletal muscle and remote organ damage affecting outcomes after total knee arthroplasty (TKA). The aim of the study is to summarize the current findings associated with I/R injury following TKA using a thigh TQ, which include cellular alterations and protective therapeutic interventions. The PubMed database was searched using the keywords "ischemia reperfusion injury," "oxidative stress," "tourniquet," and "knee arthroplasty." The search was limited to research articles published in the English language. Twenty-eight clinical studies were included in this qualitative review. Skeletal muscle I/R reduces protein synthesis, increases protein degradation, and upregulates genes in cell stress pathways. The I/R of the lower extremity elevates local and systemic oxidative stress as well as inflammatory reactions and impairs renal function. Propofol reduces oxidative injury in this I/R model. Ischemic preconditioning (IPC) and vitamin C may prevent oxygen free radical production. However, a high dose of N-acetylcysteine possibly induces kidney injury. In summary, TQ-related I/R during TKA leads to muscle protein metabolism alteration, endothelial dysfunction, oxidative stress, inflammatory response, and renal function disturbance. Propofol, IPC, and vitamin C show protective effects on oxidative and inflammatory markers. However, a relationship between biochemical parameters and postoperative clinical outcomes has not been validated.
Collapse
|
20
|
Rosenberg JH, Werner JH, Moulton MJ, Agrawal DK. Current Modalities and Mechanisms Underlying Cardioprotection by Ischemic Conditioning. J Cardiovasc Transl Res 2018; 11:292-307. [PMID: 29797232 PMCID: PMC6117210 DOI: 10.1007/s12265-018-9813-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023]
Abstract
Ischemic preconditioning is a process which serves to mitigate reperfusion injury. Preconditioning of the heart can be achieved through natural, pharmacological, and mechanical means. Mechanical preconditioning appears to have the greatest chance of good outcomes while methods employing pharmacologic preconditioning have been largely unsuccessful. Remote ischemic preconditioning achieves a cardioprotective effect by applying cycles of ischemia and reperfusion in a distal limb, stimulating the release of a neurohumoral cardioprotective factor incited by stimulation of afferent neurons. The cardioprotective factor stimulates the reperfusion injury salvage kinase (RISK) and survivor activator factor enhancement (SAFE) signaling cascades in cardiomyocytes which promote cell survival by the expression of anti-apoptotic genes and inhibition of the opening of mitochondrial permeability transition pores. Clinical application of ischemic preconditioning involving targets in the RISK and SAFE signaling appears promising in the treatment of acute myocardial infarction; however, clinical trials have yet to demonstrate additional benefit to current therapy.
Collapse
Affiliation(s)
- John H Rosenberg
- Department of Clinical & Translational Science, The Peekie Nash Carpenter Endowed Chair in Medicine, Creighton University School of Medicine, CRISS II Room 510, 2500 California Plaza, Omaha, NE, 68178, USA
| | - John H Werner
- Department of Clinical & Translational Science, The Peekie Nash Carpenter Endowed Chair in Medicine, Creighton University School of Medicine, CRISS II Room 510, 2500 California Plaza, Omaha, NE, 68178, USA
| | - Michael J Moulton
- Department of Cardiothoracic Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Devendra K Agrawal
- Department of Clinical & Translational Science, The Peekie Nash Carpenter Endowed Chair in Medicine, Creighton University School of Medicine, CRISS II Room 510, 2500 California Plaza, Omaha, NE, 68178, USA.
| |
Collapse
|
21
|
Zhang M, Gu WW, Hong XY. Involvement of Endothelin 1 in Remote Preconditioning-Induced Cardioprotection through connexin 43 and Akt/GSK-3β Signaling Pathway. Sci Rep 2018; 8:10941. [PMID: 30026513 PMCID: PMC6053397 DOI: 10.1038/s41598-018-29196-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 07/04/2018] [Indexed: 01/19/2023] Open
Abstract
The present study was aimed to explore the role of endothelins in remote preconditioning (RP)-induced myocardial protection in ischemia-reperfusion (IR) injury. RP stimulus was given by subjecting hind limb to four cycles of ischemia and reperfuion (5 minutes each) using blood pressure cuff in male rats. Following RP, hearts were isolated and subjected to 30 minutes of ischemia and 120 minutes of reperfusion on Langendorff apparatus. The extent of myocardial injury was determined by measuring the levels of LDH-1, CK-MB and cardiac troponin T (cTnT) in coronary effluent; caspase-3 activity and Bcl 2 expression in heart (apoptosis); infarct size by triphenyl tetrazolium chloride and contractility parameters including left ventricular developed pressure, dp/dtmax dp/dtmin and heart rate. RP reduced ischemia reperfusion-induced myocardial injury, increased the levels of endothelin 1 (in blood), Akt-P, GSK-3β-P and P-connexin 43 (in hearts). Pretreatment with ETA receptor antagonist, BQ 123 (1 and 2 mg/kg), ETB receptor antagonist, BQ 788 (1 and 3 mg/kg) and dual inhibitor of ETA and ETB receptor, bonsentan (25 and 50 mg/kg) abolished these effects of RP. However, the effects of bonsentan were more pronounced in comparison to BQ 123 and BQ 788. It is concluded that RP stimulus may release endothelin 1 in the blood, which may activate myocardial ETA and ETB receptors to trigger cardioprotection through connexin 43 and Akt/GSK-3β pathway.
Collapse
Affiliation(s)
- Min Zhang
- Hepatobiliary pancreatic surgery, China-Japan Union Hospital of Jilin University, 126 XianTaiStreet, Changchun, 130033, China
| | - Wei Wei Gu
- Hepatobiliary pancreatic surgery, China-Japan Union Hospital of Jilin University, 126 XianTaiStreet, Changchun, 130033, China
| | - Xing Yu Hong
- Vascular surgery, China-Japan Union Hospital of Jilin University, 126 XianTai Street, Changchun, 130033, China.
| |
Collapse
|
22
|
Chen K, Xu Z, Liu Y, Wang Z, Li Y, Xu X, Chen C, Xia T, Liao Q, Yao Y, Zeng C, He D, Yang Y, Tan T, Yi J, Zhou J, Zhu H, Ma J, Zeng C. Irisin protects mitochondria function during pulmonary ischemia/reperfusion injury. Sci Transl Med 2018; 9:9/418/eaao6298. [PMID: 29187642 DOI: 10.1126/scitranslmed.aao6298] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 08/10/2017] [Accepted: 10/20/2017] [Indexed: 12/13/2022]
Abstract
Limb remote ischemic preconditioning (RIPC) is an effective means of protection against ischemia/reperfusion (IR)-induced injury to multiple organs. Many studies are focused on identifying endocrine mechanisms that underlie the cross-talk between muscle and RIPC-mediated organ protection. We report that RIPC releases irisin, a myokine derived from the extracellular portion of fibronectin domain-containing 5 protein (FNDC5) in skeletal muscle, to protect against injury to the lung. Human patients with neonatal respiratory distress syndrome show reduced concentrations of irisin in the serum and increased irisin concentrations in the bronchoalveolar lavage fluid, suggesting transfer of irisin from circulation to the lung under physiologic stress. In mice, application of brief periods of ischemia preconditioning stimulates release of irisin into circulation and transfer of irisin to the lung subjected to IR injury. Irisin, via lipid raft-mediated endocytosis, enters alveolar cells and targets mitochondria. Interaction between irisin and mitochondrial uncoupling protein 2 (UCP2) allows for prevention of IR-induced oxidative stress and preservation of mitochondrial function. Animal model studies show that intravenous administration of exogenous irisin protects against IR-induced injury to the lung via improvement of mitochondrial function, whereas in UCP2-deficient mice or in the presence of a UCP2 inhibitor, the protective effect of irisin is compromised. These results demonstrate that irisin is a myokine that facilitates RIPC-mediated lung protection. Targeting the action of irisin in mitochondria presents a potential therapeutic intervention for pulmonary IR injury.
Collapse
Affiliation(s)
- Ken Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China.,Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Zaicheng Xu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Yukai Liu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Zhen Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Yu Li
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Xuefei Xu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Caiyu Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Tianyang Xia
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Qiao Liao
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Yonggang Yao
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Cindy Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Duofen He
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Yongjian Yang
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Tao Tan
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jianxun Yi
- Department of Physiology, Kansas City University, Kansas City, MO 64106, USA
| | - Jingsong Zhou
- Department of Physiology, Kansas City University, Kansas City, MO 64106, USA
| | - Hua Zhu
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jianjie Ma
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA.
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China. .,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| |
Collapse
|
23
|
Abdul-Ghani S, Fleishman AN, Khaliulin I, Meloni M, Angelini GD, Suleiman MS. Remote ischemic preconditioning triggers changes in autonomic nervous system activity: implications for cardioprotection. Physiol Rep 2018; 5:5/3/e13085. [PMID: 28193783 PMCID: PMC5309573 DOI: 10.14814/phy2.13085] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 11/21/2016] [Accepted: 11/22/2016] [Indexed: 01/30/2023] Open
Abstract
Cardioprotective efficacy of remote ischemic preconditioning (RIPC) remains controversial. Experimental studies investigating RIPC have largely monitored cardiovascular changes during index ischemia and reperfusion with little work investigating changes during RIPC application. This work aims to identify cardiovascular changes associated with autonomic nervous system (ANS) activity during RIPC and prior to index ischemia. RIPC was induced in anesthetized male C57/Bl6 mice by four cycles of 5 min of hindlimb ischemia using inflated cuff (200 mmHg) followed by 5 min reperfusion. Electrocardiography (ECG) and microcirculatory blood flow in both hindlimbs were recorded throughout RIPC protocol. Heart rate variability (HRV) analysis was performed using ECG data. Hearts extracted at the end of RIPC protocol were used either for measurement of myocardial metabolites using high‐performance liquid chromatography or for Langendorff perfusion to monitor function and injury during 30 min index ischemia and 2 h reperfusion. Isolated‐perfused hearts from RIPC animals had significantly less infarct size after index ischemia and reperfusion (34 ± 5% vs. 59 ± 7%; mean ± SE P < 0.05). RIPC protocol was associated with increased heart rate measured both in ex vivo and in vivo. Frequency ratio of HRV spectra was altered in RIPC compared to control. RIPC was associated with a standard hyperemic response in the cuffed‐limb but there was a sustained reduction in blood flow in the uncuffed contralateral limb. RIPC hearts (prior to index ischemia) had significantly lower phosphorylation potential and energy charge compared to the control group. In conclusion, RIPC is associated with changes in ANS activity (heart rate, blood flow, HRV) and mild myocardial ischemic stress that would contribute to cardioprotection.
Collapse
Affiliation(s)
- Safa Abdul-Ghani
- Bristol Heart Institute, School of Clinical Sciences, Faculty of Medicine & Dentistry, University of Bristol, Bristol, United Kingdom
| | - Arnold N Fleishman
- Research Institute for Complex Problems of Hygiene and Occupational Diseases, Novokuznetsk Kemerovo Oblast, Russia
| | - Igor Khaliulin
- Bristol Heart Institute, School of Clinical Sciences, Faculty of Medicine & Dentistry, University of Bristol, Bristol, United Kingdom
| | - Marco Meloni
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Gianni D Angelini
- Bristol Heart Institute, School of Clinical Sciences, Faculty of Medicine & Dentistry, University of Bristol, Bristol, United Kingdom
| | - M-Saadeh Suleiman
- Bristol Heart Institute, School of Clinical Sciences, Faculty of Medicine & Dentistry, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
24
|
De Smet S, D'Hulst G, Poffé C, Van Thienen R, Berardi E, Hespel P. High-intensity interval training in hypoxia does not affect muscle HIF responses to acute hypoxia in humans. Eur J Appl Physiol 2018; 118:847-862. [PMID: 29423544 DOI: 10.1007/s00421-018-3820-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 01/31/2018] [Indexed: 12/14/2022]
Abstract
PURPOSE The myocellular response to hypoxia is primarily regulated by hypoxia-inducible factors (HIFs). HIFs thus conceivably are implicated in muscular adaptation to altitude training. Therefore, we investigated the effect of hypoxic versus normoxic training during a period of prolonged hypoxia ('living high') on muscle HIF activation during acute ischaemia. METHODS Ten young male volunteers lived in normobaric hypoxia for 5 weeks (5 days per week, ~ 15.5 h per day, FiO2: 16.4-14.0%). One leg was trained in hypoxia (TRHYP, 12.3% FiO2) whilst the other leg was trained in normoxia (TRNOR, 20.9% FiO2). Training sessions (3 per week) consisted of intermittent unilateral knee extensions at 20-25% of the 1-repetition maximum. Before and after the intervention, a 10-min arterial occlusion and reperfusion of the leg was performed. Muscle oxygenation status was continuously measured by near-infrared spectroscopy. Biopsies were taken from m. vastus lateralis before and at the end of the occlusion. RESULTS Irrespective of training, occlusion elevated the fraction of HIF-1α expressing myonuclei from ~ 54 to ~ 64% (P < 0.05). However, neither muscle HIF-1α or HIF-2α protein abundance, nor the expression of HIF-1α or downstream targets selected increased in any experimental condition. Training in both TRNOR and TRHYP raised muscular oxygen extraction rate upon occlusion by ~ 30%, whilst muscle hyperperfusion immediately following the occlusion increased by ~ 25% in either group (P < 0.05). CONCLUSION Ten minutes of arterial occlusion increased HIF-1α-expressing myonuclei. However, neither normoxic nor hypoxic training during 'living high' altered muscle HIF translocation, stabilisation, or transcription in response to acute hypoxia induced by arterial occlusion.
Collapse
Affiliation(s)
- Stefan De Smet
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Tervuursevest 101, 3001, Leuven, Belgium
| | - Gommaar D'Hulst
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Tervuursevest 101, 3001, Leuven, Belgium.,Laboratory of Exercise and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Chiel Poffé
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Tervuursevest 101, 3001, Leuven, Belgium
| | - Ruud Van Thienen
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Tervuursevest 101, 3001, Leuven, Belgium
| | - Emanuele Berardi
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Tervuursevest 101, 3001, Leuven, Belgium
| | - Peter Hespel
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Tervuursevest 101, 3001, Leuven, Belgium. .,Bakala Academy-Athletic Performance Center, KU Leuven, Leuven, Belgium.
| |
Collapse
|
25
|
de Preux Charles AS, Bise T, Baier F, Sallin P, Jaźwińska A. Preconditioning boosts regenerative programmes in the adult zebrafish heart. Open Biol 2017; 6:rsob.160101. [PMID: 27440423 PMCID: PMC4967829 DOI: 10.1098/rsob.160101] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/20/2016] [Indexed: 12/22/2022] Open
Abstract
During preconditioning, exposure to a non-lethal harmful stimulus triggers a body-wide increase of survival and pro-regenerative programmes that enable the organism to better withstand the deleterious effects of subsequent injuries. This phenomenon has first been described in the mammalian heart, where it leads to a reduction of infarct size and limits the dysfunction of the injured organ. Despite its important clinical outcome, the actual mechanisms underlying preconditioning-induced cardioprotection remain unclear. Here, we describe two independent models of cardiac preconditioning in the adult zebrafish. As noxious stimuli, we used either a thoracotomy procedure or an induction of sterile inflammation by intraperitoneal injection of immunogenic particles. Similar to mammalian preconditioning, the zebrafish heart displayed increased expression of cardioprotective genes in response to these stimuli. As zebrafish cardiomyocytes have an endogenous proliferative capacity, preconditioning further elevated the re-entry into the cell cycle in the intact heart. This enhanced cycling activity led to a long-term modification of the myocardium architecture. Importantly, the protected phenotype brought beneficial effects for heart regeneration within one week after cryoinjury, such as a more effective cell-cycle reentry, enhanced reactivation of embryonic gene expression at the injury border, and improved cell survival shortly after injury. This study reveals that exposure to antecedent stimuli induces adaptive responses that render the fish more efficient in the activation of the regenerative programmes following heart damage. Our results open a new field of research by providing the adult zebrafish as a model system to study remote cardiac preconditioning.
Collapse
Affiliation(s)
| | - Thomas Bise
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Felix Baier
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Pauline Sallin
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| |
Collapse
|
26
|
José Alburquerque-Béjar J, Barba I, Valls-Lacalle L, Ruiz-Meana M, Pecoraro M, Rodríguez-Sinovas A, García-Dorado D. Remote ischemic conditioning provides humoural cross-species cardioprotection through glycine receptor activation. Cardiovasc Res 2017; 113:52-60. [PMID: 28069702 DOI: 10.1093/cvr/cvw242] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/18/2016] [Accepted: 12/02/2016] [Indexed: 11/12/2022] Open
Abstract
AIMS Remote ischaemic conditioning (RIC) releases a humoural factor able to exert cross-species cardioprotection when plasma dialysate is applied to isolated hearts. However, the exact chemical nature of this factor is currently unknown. METHODS AND RESULTS RIC (4 × 5min femoral occlusion/5min reperfusion) was applied to 10 male pigs, and blood was taken before and after the manoeuvre. Discriminant analysis of 1H-NMR spectra (n = 10-12) obtained from plasma dialysates (12-14 kDa cut-off) allowed to demonstrate a different metabolic profile between control and postRIC samples, with lactate (2.671 ± 0.294 vs. 3.666 ± 0.291 μmol/mL, P = 0.020), succinate (0.062 ± 0.005 vs. 0.082 ± 0.008 μmol/mL, P = 0.035) and glycine (0.055 ± 0.009 vs. 0.471 ± 0.151 μmol/mL, P = 0.015) being the main responsible for such differences. Plasma dialysates were then given to isolated mice hearts submitted to global ischaemia (35 min) and reperfusion (60 min), for 30 min before ischaemia or during the first 15 min of reflow. Infarct size was significantly reduced when postRIC dialysate was applied before ischaemia as compared with hearts pretreated with control dialysate (44.81 ± 3.22 vs. 55.55 ± 2.53%, P = 0.012, n = 12). Blockade of glycine receptors with strychnine 10 μM inhibited the protective effect caused by pretreatment with postRIC dialysate (52.76 ± 6.94 vs. 51.92 ± 5.78%, P-NS, n = 5), whereas pretreatment with glycine 3 mmol/L, but not succinate 100 μmol/L, mimicked RIC protection (41.90 ± 4.50% in glycine-treated vs. 61.51 ± 5.16 and 64.73 ± 4.47% in succinate-treated and control hearts, respectively, P < 0.05, n = 4-7). CONCLUSIONS RIC releases glycine and exerts cross-species cardioprotection against infarction through glycine receptor activation.
Collapse
Affiliation(s)
- Juan José Alburquerque-Béjar
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Ignasi Barba
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Laura Valls-Lacalle
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Marisol Ruiz-Meana
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Michela Pecoraro
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Antonio Rodríguez-Sinovas
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - David García-Dorado
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg Vall d'Hebron 119-129, 08035 Barcelona, Spain
| |
Collapse
|
27
|
Abstract
Part I of this review discussed the similarities between embryogenesis, mammalian adaptions to hypoxia (primarily driven by hypoxia-inducible factor-1 [HIF-1]), ischemia-reperfusion injury (and its relationship with reactive oxygen species), hibernation, diving animals, cancer, and sepsis, and it focused on the common characteristics that allow cells and organisms to survive in these states. Part II of this review describes techniques by which researchers gain insight into subcellular energetics and identify potential future tools for clinicians. In particular, P nuclear magnetic resonance to measure high-energy phosphates, serum lactate measurements, the use of near-infrared spectroscopy to measure the oxidation state of cytochrome aa3, and the ability of the protoporphyrin IX-triplet state lifetime technique to measure mitochondrial oxygen tension are discussed. In addition, this review discusses novel treatment strategies such as hyperbaric oxygen, preconditioning, exercise training, therapeutic gases, as well as inhibitors of HIF-1, HIF prolyl hydroxylase, and peroxisome proliferator-activated receptors.
Collapse
Affiliation(s)
- Robert H Thiele
- From the Department of Anesthesiology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
28
|
Remote Ischemic Preconditioning: A Novel Strategy in Rescuing Older Livers From Ischemia-reperfusion Injury in a Rodent Model. Ann Surg 2017; 264:797-803. [PMID: 27584570 DOI: 10.1097/sla.0000000000001765] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The aim of this study was to determine whether remote ischemic preconditioning (RIPC) protects aged liver against ischemia reperfusion (IR). SUMMARY OF BACKGROUND DATA The demands for liver surgery in an aging population are growing. Clamping of vessels to prevent blood loss is integral to liver surgery, but the resulting IR injury (IRI) augments postoperative complications. More so, sensitivity to hepatic IRI increases with age; however, no strategies have been developed that specifically protect old liver. RIPC, a novel protective approach, was performed distant to the surgical site. Whether RIPC may also protect old liver from IRI is unknown. METHODS RIPC to the femoral vascular bundle was compared against direct ischemic preconditioning (IPC) and the standard of care intermittent clamping (IC) using a model of partial hepatic ischemia in mice aged 20 to 24 months. Liver injury was measured 6 hours after reperfusion. Protective signaling (serotonin-Vegf-Il10/Mmp8 axis, Kupffer cell polarization) was assessed immediately after preconditioning. Neutralizing antibody was used to test the role of Vegf. Hepatic vasculature was examined by electron microscopy. RESULTS RIPC was superior over other strategies in protecting old liver from IRI, with standard IPC approaches being ineffective. RIPC induced the strongest elevations in circulating Vegf, and Vegf inhibition dampened protective signaling and abrogated the protective effects. RIPC was further associated with improvements in vascular functionality. CONCLUSIONS RIPC is highly effective in protecting old liver from ischemic insults, mainly owing to its ability to induce circulating Vegf. These findings warrant efforts toward clinical translation.
Collapse
|
29
|
Herajärvi J, Anttila T, Dimova EY, Laukka T, Myllymäki M, Haapanen H, Olenchock BA, Tuominen H, Puistola U, Karihtala P, Kiviluoma K, Koivunen P, Anttila V, Juvonen T. Exploring effects of remote ischemic preconditioning in a pig model of hypothermic circulatory arrest. SCAND CARDIOVASC J 2017; 51:233-241. [PMID: 28434264 DOI: 10.1080/14017431.2017.1319574] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVES During aortic and cardiac surgery, risks for mortality and morbidity are inevitable. Surgical setups involving deep hypothermic circulatory arrest (DHCA) are effective to achieve organ protection against ischemic injury. The aim of this study was to identify humoural factors mediating additive protective effects of remote ischemic preconditioning (RIPC) in a porcine model of DHCA. DESIGN Twenty-two pigs were randomized into the RIPC group (n = 11) and the control group (n = 11). The RIPC group underwent four 5-minute hind limb ischemia-reperfusion cycles prior to cardiopulmonary bypass and DHCA. All animals underwent identical surgical procedures including 60 min DHCA at 18 °C. Blood samples were collected from vena cava and sagittal sinus at several time points. After the 8-hour follow-up period, the brain, heart, and kidney tissue samples were collected for tissue analyses. RESULTS Serum levels of brain damage marker S100B recovered faster in the RIPC group, after 4 hours of the arrest, (p < .05). Systemic lactate levels were lower and cardiac index was higher in the RIPC group postoperatively. Immunohistochemical cerebellum regional scores of antioxidant response regulator Nrf2 were better in the RIPC group (mean: 1.1, IQR: 0.0-2.5) compared with the control group (mean: 0.0, IQR: 0.0-0.0), reaching borderline statistical significance (p = .064). RIPC induced detectable modulations of plasma proteome and metabolites. CONCLUSIONS The faster recovery of S100B, lower systemic lactate levels and favourable regional antioxidant response suggest possible neuronal cellular and mitochondrial protection by RIPC, whereas better cardiac index underlines functional effects of RIPC. The exact humoural factor remains unclear.
Collapse
Affiliation(s)
- Johanna Herajärvi
- a Research Unit of Surgery, Anesthesia and Intensive Care , University of Oulu and MRC Oulu , Oulu, Finland
| | - Tuomas Anttila
- a Research Unit of Surgery, Anesthesia and Intensive Care , University of Oulu and MRC Oulu , Oulu, Finland
| | - Elitsa Y Dimova
- b Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine , Oulu Center for Cell-Matrix Research, University of Oulu , Oulu , Finland
| | - Tuomas Laukka
- b Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine , Oulu Center for Cell-Matrix Research, University of Oulu , Oulu , Finland
| | - Mikko Myllymäki
- b Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine , Oulu Center for Cell-Matrix Research, University of Oulu , Oulu , Finland
| | - Henri Haapanen
- a Research Unit of Surgery, Anesthesia and Intensive Care , University of Oulu and MRC Oulu , Oulu, Finland
| | - Benjamin A Olenchock
- c Division of Cardiovascular Medicine, Department of Medicine, The Brigham and Women's Hospital , Harvard Medical School , Boston , MA , USA
| | - Hannu Tuominen
- d Department of Pathology , MRC Oulu, Oulu University Hospital and University of Oulu , Oulu , Finland
| | - Ulla Puistola
- e Department of Obstetrics and Gynaecology , MRC Oulu, Oulu University Hospital and University of Oulu , Oulu , Finland
| | - Peeter Karihtala
- f Department of Oncology and Radiotherapy , MRC Oulu, Oulu University Hospital and University of Oulu , Oulu , Finland
| | - Kai Kiviluoma
- a Research Unit of Surgery, Anesthesia and Intensive Care , University of Oulu and MRC Oulu , Oulu, Finland
| | - Peppi Koivunen
- b Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine , Oulu Center for Cell-Matrix Research, University of Oulu , Oulu , Finland
| | - Vesa Anttila
- a Research Unit of Surgery, Anesthesia and Intensive Care , University of Oulu and MRC Oulu , Oulu, Finland.,g Heart Center , Turku University Hospital, University of Turku , Turku , Finland
| | - Tatu Juvonen
- a Research Unit of Surgery, Anesthesia and Intensive Care , University of Oulu and MRC Oulu , Oulu, Finland.,h Department of Cardiac Surgery , HUCH Heart and Lung Center , Helsinki , Finland
| |
Collapse
|
30
|
Ravingerova T, Farkasova V, Griecsova L, Carnicka S, Murarikova M, Barlaka E, Kolar F, Bartekova M, Lonek L, Slezak J, Lazou A. Remote preconditioning as a novel "conditioning" approach to repair the broken heart: potential mechanisms and clinical applications. Physiol Res 2017; 65 Suppl 1:S55-64. [PMID: 27643940 DOI: 10.33549/physiolres.933392] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Remote ischemic preconditioning (RIPC) is a novel strategy of protection against ischemia-reperfusion (IR) injury in the heart (and/or other organs) by brief episodes of non-lethal IR in a distant organ/tissue. Importantly, RIPC can be induced noninvasively by limitation of blood flow in the extremity implying the applicability of this method in clinical situations. RIPC (and its delayed phase) is a form of relatively short-term adaptation to ischemia, similar to ischemic PC, and likely they both share triggering mechanisms, whereas mediators and end-effectors may differ. It is hypothesized that communication between the signals triggered in the remote organs and protection in the target organ may be mediated through substances released from the preconditioned organ and transported via the circulation (humoral pathways), by neural pathways and/or via systemic anti-inflammatory and antiapoptotic response to short ischemic bouts. Identification of molecules involved in RIPC cascades may have therapeutic and diagnostic implications in the management of myocardial ischemia. Elucidation of the mechanisms of endogenous cardioprotection triggered in the remote organ could lead to the development of diverse pharmacological RIPC mimetics. In the present article, the authors provide a short overview of RIPC-induced protection, proposed underlying mechanisms and factors modulating RIPC as a promising cardioprotective strategy.
Collapse
Affiliation(s)
- T Ravingerova
- Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Aspectos perioperatorios: riesgo quirúrgico y fragilidad, condicionamiento isquémico remoto, umbral transfusional, postoperatorio inmediato y vías de corta estancia. CIRUGIA CARDIOVASCULAR 2017. [DOI: 10.1016/j.circv.2017.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
32
|
Feng J, Yang Y, Zhou Y, Wang B, Xiong H, Fan C, Jiang S, Liu J, Ma Z, Hu W, Li T, Feng X, Xu J, Jin Z. Bakuchiol attenuates myocardial ischemia reperfusion injury by maintaining mitochondrial function: the role of silent information regulator 1. Apoptosis 2016; 21:532-45. [PMID: 27000151 DOI: 10.1007/s10495-016-1225-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Ischemia reperfusion (IR) injury (IRI) is associated with poor prognoses in the settings of both cardiac surgery and ischemic heart disease and causes mitochondrial oxidative stress and cell death. Silent information regulator 1 (SIRT1), a member of the histone deacetylase family, exerts anti-IRI effects. Bakuchiol (BAK), an analog of resveratrol and a monoterpene phenol isolated from the seeds of Psoralea corylifolia (Leguminosae), protects tissues from injury. This study was designed to investigate the protective effects of BAK treatment in the setting of myocardial IRI and to elucidate the potential mechanism of those effects. Prior to induction of IR, isolated rat hearts or cardiomyocytes were exposed to BAK in either the absence or presence of the SIRT1 inhibitors Sirtinol and SIRT1 siRNA. BAK exerted cardioprotective effects, as evidenced by the improvements noted in cardiac function following ischemia, attenuated myocardial apoptosis, and changes in several biochemical parameters (including increases in the level of the anti-apoptotic protein Bcl2, decreases in the level of the pro-apoptotic protein Bax, and decreases in the cleaved Caspase 3 level). However, Sirtinol and SIRT1 siRNA each blocked BAK-induced cardioprotection by inhibiting SIRT1 signaling. Additionally, BAK significantly increased the activities of mitochondrial succinate dehydrogenase, cytochrome c oxidase, and mitochondrial superoxide dismutase and decreased the production of malondialdehyde. These findings suggested that BAK significantly attenuated IR-induced mitochondrial oxidative damage. However, Sirtinol and SIRT1 siRNA abolished BAK-dependent mitochondrial function. In summary, our results demonstrate that BAK treatment attenuates IRI by attenuating IR-induced mitochondrial oxidative damage via the activation of SIRT1/PGC-1α signaling.
Collapse
Affiliation(s)
- Jianyu Feng
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Yang Yang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Yajun Zhou
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Department of Cardiothoracic Surgery, The 94th Hospital of Chinese PLA, 1028 Jingangshan Road, Nanchang, 330000, China
| | - Bodong Wang
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Hongyan Xiong
- Department of Cardiothoracic Surgery, Central Hospital of Xi'an, 185 Houzaimen Road, Xi'an, 710033, China
| | - Chongxi Fan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710038, China
| | - Shuai Jiang
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Jun Liu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710038, China
| | - Wei Hu
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Xiao Feng
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Jianjun Xu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China.
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, China.
| | - Zhenxiao Jin
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
33
|
Frumkin K, Bloom AS. Ischemic Conditioning: Implications for Emergency Medicine. Ann Emerg Med 2016; 68:268-74. [DOI: 10.1016/j.annemergmed.2016.02.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 02/08/2016] [Accepted: 02/10/2016] [Indexed: 01/22/2023]
|
34
|
Samanta A, Dawn B. Remote Ischemic Preconditioning for Cardiac Surgery: Reflections on Evidence of Efficacy. Circ Res 2016; 118:1055-8. [PMID: 27034273 DOI: 10.1161/circresaha.116.308373] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Anweshan Samanta
- From the Division of Cardiovascular Diseases, Cardiovascular Research Institute, The Midwest Stem Cell Therapy Center, University of Kansas Medical Center and Hospital, Kansas City
| | - Buddhadeb Dawn
- From the Division of Cardiovascular Diseases, Cardiovascular Research Institute, The Midwest Stem Cell Therapy Center, University of Kansas Medical Center and Hospital, Kansas City.
| |
Collapse
|
35
|
Reconciling the IPC and Two-Hit Models: Dissecting the Underlying Cellular and Molecular Mechanisms of Two Seemingly Opposing Frameworks. J Immunol Res 2015; 2015:697193. [PMID: 26770993 PMCID: PMC4684872 DOI: 10.1155/2015/697193] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/10/2015] [Accepted: 11/18/2015] [Indexed: 12/30/2022] Open
Abstract
Inflammatory cascades and mechanisms are ubiquitous during host responses to various types of insult. Biological models and interventional strategies have been devised as an effort to better understand and modulate inflammation-driven injuries. Amongst those the two-hit model stands as a plausible and intuitive framework that explains some of the most frequent clinical outcomes seen in injuries like trauma and sepsis. This model states that a first hit serves as a priming event upon which sequential insults can build on, culminating on maladaptive inflammatory responses. On a different front, ischemic preconditioning (IPC) has risen to light as a readily applicable tool for modulating the inflammatory response to ischemia and reperfusion. The idea is that mild ischemic insults, either remote or local, can cause organs and tissues to be more resilient to further ischemic insults. This seemingly contradictory role that the two models attribute to a first inflammatory hit, as priming in the former and protective in the latter, has set these two theories on opposing corners of the literature. The present review tries to reconcile both models by showing that, rather than debunking each other, each framework offers unique insights in understanding and modulating inflammation-related injuries.
Collapse
|
36
|
Sharma R, Randhawa PK, Singh N, Jaggi AS. Possible role of thromboxane A2 in remote hind limb preconditioning-induced cardioprotection. Naunyn Schmiedebergs Arch Pharmacol 2015; 389:1-9. [DOI: 10.1007/s00210-015-1186-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 10/23/2015] [Indexed: 01/10/2023]
|
37
|
Aimo A, Borrelli C, Giannoni A, Pastormerlo LE, Barison A, Mirizzi G, Emdin M, Passino C. Cardioprotection by remote ischemic conditioning: Mechanisms and clinical evidences. World J Cardiol 2015; 7:621-632. [PMID: 26516416 PMCID: PMC4620073 DOI: 10.4330/wjc.v7.i10.621] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 06/29/2015] [Accepted: 08/14/2015] [Indexed: 02/06/2023] Open
Abstract
In remote ischemic conditioning (RIC), several cycles of ischemia and reperfusion render distant organ and tissues more resistant to the ischemia-reperfusion injury. The intermittent ischemia can be applied before the ischemic insult in the target site (remote ischemic preconditioning), during the ischemic insult (remote ischemic perconditioning) or at the onset of reperfusion (remote ischemic postconditioning). The mechanisms of RIC have not been completely defined yet; however, these mechanisms must be represented by the release of humoral mediators and/or the activation of a neural reflex. RIC has been discovered in the heart, and has been arising great enthusiasm in the cardiovascular field. Its efficacy has been evaluated in many clinical trials, which provided controversial results. Our incomplete comprehension of the mechanisms underlying the RIC could be impairing the design of clinical trials and the interpretation of their results. In the present review we summarize current knowledge about RIC pathophysiology and the data about its cardioprotective efficacy.
Collapse
|
38
|
Waltz P, Escobar D, Botero AM, Zuckerbraun BS. Nitrate/Nitrite as Critical Mediators to Limit Oxidative Injury and Inflammation. Antioxid Redox Signal 2015; 23:328-39. [PMID: 26140517 PMCID: PMC4692126 DOI: 10.1089/ars.2015.6256] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 06/30/2015] [Accepted: 07/01/2015] [Indexed: 12/31/2022]
Abstract
SIGNIFICANCE Nitric oxide (NO) is a critical signaling molecule marked by complex chemistry and varied biological responses depending on the context of the redox environment. In the setting of inflammation, NO can not only contribute to tissue injury and be causative of oxidative damage but can also signal as an adaptive molecule to limit inflammatory signaling in multiple cell types and tissues. RECENT ADVANCES An advance in our understanding of NO biology was the recognition of the nitrate-nitrite-NO axis, whereby nitrate (predominantly from dietary sources) could be converted to nitrite and nitrite could be reduced to NO. CRITICAL ISSUES Intriguingly, the recognition of multiple enzymes that serve as nitrite reductases in the setting of hypoxia or ischemia established the concept of nitrite as a circulating endocrine reservoir of NO, with the selective release of NO at sites that were primed for this reaction. This review highlights the anti-inflammatory roles of nitrite in numerous clinical conditions, including ischemia/reperfusion, transplant, cardiac arrest, and vascular injury, and in gastrointestinal inflammation. FUTURE DIRECTIONS These preclinical and clinical investigations set up further clinical trials and studies that elucidate the endogenous role this pathway plays in protection against inflammatory signaling.
Collapse
Affiliation(s)
- Paul Waltz
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Daniel Escobar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ana Maria Botero
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Brian S. Zuckerbraun
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- The Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| |
Collapse
|
39
|
Berger M, Nadler JW, Browndyke J, Terrando N, Ponnusamy V, Cohen HJ, Whitson HE, Mathew JP. Postoperative Cognitive Dysfunction: Minding the Gaps in Our Knowledge of a Common Postoperative Complication in the Elderly. Anesthesiol Clin 2015; 33:517-50. [PMID: 26315636 DOI: 10.1016/j.anclin.2015.05.008] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Postoperative cognitive dysfunction (POCD) is a common complication associated with significant morbidity and mortality in elderly patients. There is much interest in and controversy about POCD, reflected partly in the increasing number of articles published on POCD recently. Recent work suggests surgery may also be associated with cognitive improvement in some patients, termed postoperative cognitive improvement (POCI). As the number of surgeries performed worldwide approaches 250 million per year, optimizing postoperative cognitive function and preventing/treating POCD are major public health issues. In this article, we review the literature on POCD and POCI, and discuss current research challenges in this area.
Collapse
Affiliation(s)
- Miles Berger
- Department of Anesthesiology, Duke University Medical Center, Duke South, Orange Zone, Room 4317, Durham, NC 27710, USA.
| | - Jacob W Nadler
- Department of Anesthesiology, Duke University Medical Center, Duke South, Orange Zone, Room 4317, Durham, NC 27710, USA
| | - Jeffrey Browndyke
- Department of Anesthesiology, Duke University Medical Center, Duke South, Orange Zone, Room 4317, Durham, NC 27710, USA
| | - Niccolo Terrando
- Department of Anesthesiology, Duke University Medical Center, Duke South, Orange Zone, Room 4317, Durham, NC 27710, USA
| | - Vikram Ponnusamy
- Department of Anesthesiology, Duke University Medical Center, Duke South, Orange Zone, Room 4317, Durham, NC 27710, USA
| | - Harvey Jay Cohen
- Department of Anesthesiology, Duke University Medical Center, Duke South, Orange Zone, Room 4317, Durham, NC 27710, USA
| | - Heather E Whitson
- Department of Anesthesiology, Duke University Medical Center, Duke South, Orange Zone, Room 4317, Durham, NC 27710, USA
| | - Joseph P Mathew
- Department of Anesthesiology, Duke University Medical Center, Duke South, Orange Zone, Room 4317, Durham, NC 27710, USA
| |
Collapse
|
40
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Extracellular vesicles and atherosclerotic disease. Cell Mol Life Sci 2015; 72:2697-708. [PMID: 25894694 PMCID: PMC11113133 DOI: 10.1007/s00018-015-1906-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 04/08/2015] [Accepted: 04/09/2015] [Indexed: 12/14/2022]
Abstract
Circulating extracellular vesicles (EVs) comprise a heterogeneous population of vesicular structures. According to the current paradigm, there are three types of EVs, including exosomes, microvesicles and apoptotic bodies, that are differentiated in their size, formation, and release mechanisms. EVs were shown to act as a 'post service' that serves a long-distance delivery of complex cellular messages. The cargo of EVs consists of a variety of biomolecules including proteins, DNA, mRNA, and non-coding RNA. In normal or pathological conditions, EVs deliver various molecules to the recipient cells. Those molecules greatly vary depending on the microenvironmental stimuli. In proinflammatory conditions such as atherosclerosis and other cardiovascular diseases, EVs derived from vascular endothelial cells, vascular smooth muscle cells, macrophages, and other circulating immune cells mainly possess proinflammatory properties. However, the capacity of circulating EVs to stably maintain and deliver a variety of biomolecules makes these microparticles to be a promising therapeutic tool for treatment of cardiovascular pathology. To date, circulating EVs were evaluated to be as a source of valuable diagnostic and prognostic biomarkers such as microRNA. Circulating EVs keep a great therapeutic potential to serve as vehicles for targeted therapy of cardiovascular diseases.
Collapse
Affiliation(s)
- Dimitry A. Chistiakov
- The Mount Sinai Community Clinical Oncology Program, Mount Sinai Comprehensive Cancer Center, Mount Sinai Medical Center, Miami Beach, FL 33140 USA
- Division of Laboratory Medicine, Department of Molecular Genetic Diagnostics and Cell Biology, Institute of Pediatrics, Research Center for Children’s Health, Moscow, 119991 Russia
| | - Alexander N. Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow, 125315 Russia
- Department of Biophysics, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991 Russia
| | - Yuri V. Bobryshev
- Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052 Australia
- School of Medicine, University of Western Sydney, Campbelltown, NSW 2560 Australia
| |
Collapse
|
41
|
Berger MM, Macholz F, Mairbäurl H, Bärtsch P. Remote ischemic preconditioning for prevention of high-altitude diseases: fact or fiction? J Appl Physiol (1985) 2015; 119:1143-51. [PMID: 26089545 DOI: 10.1152/japplphysiol.00156.2015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 06/17/2015] [Indexed: 01/14/2023] Open
Abstract
Preconditioning refers to exposure to brief episodes of potentially adverse stimuli and protects against injury during subsequent exposures. This was first described in the heart, where episodes of ischemia/reperfusion render the myocardium resistant to subsequent ischemic injury, which is likely caused by reactive oxygen species (ROS) and proinflammatory processes. Protection of the heart was also found when preconditioning was performed in an organ different from the target, which is called remote ischemic preconditioning (RIPC). The mechanisms causing protection seem to include stimulation of nitric oxide (NO) synthase, increase in antioxidant enzymes, and downregulation of proinflammatory cytokines. These pathways are also thought to play a role in high-altitude diseases: high-altitude pulmonary edema (HAPE) is associated with decreased bioavailability of NO and increased generation of ROS, whereas mechanisms causing acute mountain sickness (AMS) and high-altitude cerebral edema (HACE) seem to involve cytotoxic effects by ROS and inflammation. Based on these apparent similarities between ischemic damage and AMS, HACE, and HAPE, it is reasonable to assume that RIPC might be protective and improve altitude tolerance. In studies addressing high-altitude/hypoxia tolerance, RIPC has been shown to decrease pulmonary arterial systolic pressure in normobaric hypoxia (13% O2) and at high altitude (4,342 m). Our own results indicate that RIPC transiently decreases the severity of AMS at 12% O2. Thus preliminary studies show some benefit, but clearly, further experiments to establish the efficacy and potential mechanism of RIPC are needed.
Collapse
Affiliation(s)
- Marc Moritz Berger
- Department of Anesthesiology, Perioperative and General Critical Care Medicine, Salzburg General Hospital, Paracelsus Medical University, Salzburg, Austria; Department of Anesthesiology, University of Heidelberg, Heidelberg, Germany;
| | - Franziska Macholz
- Department of Anesthesiology, Perioperative and General Critical Care Medicine, Salzburg General Hospital, Paracelsus Medical University, Salzburg, Austria
| | - Heimo Mairbäurl
- Department of Internal Medicine VII, Division of Sports Medicine, University of Heidelberg, Heidelberg, Germany; and Translational Lung Research Center Heidelberg, German Center for Lung Research, Heidelberg, Germany
| | - Peter Bärtsch
- Department of Internal Medicine VII, Division of Sports Medicine, University of Heidelberg, Heidelberg, Germany; and
| |
Collapse
|
42
|
Zheng L, Chen J, Ma Z, Liu W, Yang F, Yang Z, Wang K, Wang X, He D, Li L. Capsaicin causes inactivation and degradation of the androgen receptor by inducing the restoration of miR-449a in prostate cancer. Oncol Rep 2015; 34:1027-34. [PMID: 26081756 DOI: 10.3892/or.2015.4055] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 05/27/2015] [Indexed: 11/06/2022] Open
Abstract
Capsaicin, a novel antitumor agent extracted from chili peppers, has been proven to induce growth inhibition in various types of cancer including prostate cancer. However, the detailed mechanisms remain largely undiscovered. In the present study, we explored the regulation of the androgen receptor (AR) by capsaicin and further researched the mechanisms of their interaction in AR-positive prostate cancer cells. In the present study, cell viability was assessed by MTT assay. Cell cycle distribution was determined using flow cytometry. Expression levels of cyclin D1, miR-449a, AR and prostate-specific antigen (PSA) were assessed by quantitative real-time polymerase chain reaction or western blot analysis. To further confirm the relationship among miR-449a, AR and prostate cancer proliferation, miR-449a was overexpressed by a lentivirus in prostate cancer cells. We discovered that capsaicin prevented tumor proliferation and cell cycle progression through inactivation and degradation of AR. We also found that restoration of miR-449a induced by capsaicin treatment resulted in the inhibition of AR signaling. Finally, we demonstrated that increased expression of miR-449a sensitized prostate cancer to capsaicin treatment. Finally, our experimental results indicated that capsaicin negatively modulates the activity of AR at the mRNA and protein levels by restoring miR-449a profiling in prostate cancer. In addition, increased expression of miR-449a may facilitate the sensitivity of prostate cancer to capsaicin treatment. Thus, capsaicin may be developed as a novel anti-AR drug for the therapy of prostate cancer.
Collapse
Affiliation(s)
- Long Zheng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Jiaqi Chen
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Zhenkun Ma
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Wei Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Fei Yang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Zhao Yang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Ke Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Xinyang Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Dalin He
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Lei Li
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| |
Collapse
|