1
|
Chaudhary M, Sharma V, Bedi O, Kaur A, Singh TG. SGK-1 Signalling Pathway is a Key Factor in Cell Survival in Ischemic Injury. Curr Drug Targets 2023; 24:1117-1126. [PMID: 37904552 DOI: 10.2174/0113894501239948231013072901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 06/11/2023] [Accepted: 09/15/2023] [Indexed: 11/01/2023]
Abstract
Serum and glucocorticoid-regulated kinases (SGK) are serine/threonine kinases that belong to AGC. The SGK-1, which responds to stress, controls a range of ion channels, cell growth, transcription factors, membrane transporters, cellular enzymes, cell survival, proliferation and death. Its expression is highly controlled by various factors such as hyperosmotic or isotonic oxidative stress, cell shrinkage, radiation, high blood sugar, neuronal injury, DNA damage, mechanical stress, thermal shock, excitement, dehydration and ischemia. The structural and functional deterioration that arises after a period of ischemia when blood flow is restored is referred to as ischemia/ reperfusion injury (I/R). The current review discusses the structure, expression, function and degradation of SGK-1 with special emphasis on the various ischemic injuries in different organs such as renal, myocardial, cerebral, intestinal and lungs. Furthermore, this review highlights the various therapeutic agents that activate the SGK-1 pathway and slow down the progression of I/R injuries.
Collapse
Affiliation(s)
- Manisha Chaudhary
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Onkar Bedi
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | | |
Collapse
|
2
|
Mayoral-González I, Calderón-Sánchez EM, Galeano-Otero I, Martín-Bórnez M, Gutiérrez-Carretero E, Fernández-Velasco M, Domenech N, Crespo-Leiro MG, Gómez AM, Ordóñez-Fernández A, Hmadcha A, Smani T. Cardiac protection induced by urocortin-2 enables the regulation of apoptosis and fibrosis after ischemia and reperfusion involving miR-29a modulation. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:838-853. [PMID: 35141045 PMCID: PMC8807986 DOI: 10.1016/j.omtn.2022.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 01/07/2022] [Indexed: 12/31/2022]
Abstract
Urocortin-2 (Ucn-2) has demonstrated cardioprotective actions against myocardial ischemia-reperfusion (I/R) injuries. Herein, we explored the protective role of Ucn-2 through microRNAs (miRNAs) post-transcriptional regulation of apoptotic and pro-fibrotic genes. We determined that the intravenous administration of Ucn-2 before heart reperfusion in a Wistar rat model of I/R recovered cardiac contractility and decreased fibrosis, lactate dehydrogenase release, and apoptosis. The infusion of Ucn-2 also inhibited the upregulation of 6 miRNAs in revascularized heart. The in silico analysis indicated that miR-29a and miR-451_1∗ are predicted to target many apoptotic and fibrotic genes. Accordingly, the transfection of neonatal rat ventricular myocytes with mimics overexpressing miR-29a, but not miR-451_1∗, prevented I/R-induced expression of pro- and anti-apoptotic genes such as Apaf-1, Hmox-1, and Cycs, as well as pro-fibrotic genes Col-I and Col-III. We also confirmed that Hmox-1, target of miR-29a, is highly expressed at the mRNA and protein levels in adult rat heart under I/R, whereas, Ucn-2 abolished I/R-induced mRNA and protein upregulation of HMOX-1. Interestingly, a significant upregulation of Hmox-1 was observed in the ventricle of ischemic patients with heart failure, correlating negatively with the left ventricle ejection fraction. Altogether, these data indicate that Ucn-2, through miR-29a regulation, provides long-lasting cardioprotection, involving the post-transcriptional regulation of apoptotic and fibrotic genes.
Collapse
Affiliation(s)
- Isabel Mayoral-González
- Department of Surgery, University of Seville, Seville, Spain
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/University of Seville/CSIC, Seville, Spain
| | - Eva M. Calderón-Sánchez
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/University of Seville/CSIC, Seville, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovaculares (CIBERCV), Madrid, Spain
| | - Isabel Galeano-Otero
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/University of Seville/CSIC, Seville, Spain
- Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain
| | - Marta Martín-Bórnez
- Department of Surgery, University of Seville, Seville, Spain
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/University of Seville/CSIC, Seville, Spain
| | - Encarnación Gutiérrez-Carretero
- Department of Surgery, University of Seville, Seville, Spain
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/University of Seville/CSIC, Seville, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovaculares (CIBERCV), Madrid, Spain
| | - María Fernández-Velasco
- Centro de Investigación Biomédica en Red Enfermedades Cardiovaculares (CIBERCV), Madrid, Spain
- Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Nieves Domenech
- Centro de Investigación Biomédica en Red Enfermedades Cardiovaculares (CIBERCV), Madrid, Spain
- Cardiology Department, Instituto de Investigación Biomédica de A Coruña, Complexo Hospitalario Universitario de A Coruña, Servicio Gallego de Salud, Universidade da Coruña, Coruña, Spain
| | - María Generosa Crespo-Leiro
- Centro de Investigación Biomédica en Red Enfermedades Cardiovaculares (CIBERCV), Madrid, Spain
- Cardiology Department, Instituto de Investigación Biomédica de A Coruña, Complexo Hospitalario Universitario de A Coruña, Servicio Gallego de Salud, Universidade da Coruña, Coruña, Spain
| | - Ana María Gómez
- Signaling and Cardiovascular Pathophysiology, INSERM, Université Paris Saclay, Châtenay-Malabry, France
| | - Antonio Ordóñez-Fernández
- Department of Surgery, University of Seville, Seville, Spain
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/University of Seville/CSIC, Seville, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovaculares (CIBERCV), Madrid, Spain
| | - Abdelkrim Hmadcha
- Department of Biotechnology, University of Alicante, Alicante, Spain
- University of Pablo Olavide, Seville, Spain
| | - Tarik Smani
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/University of Seville/CSIC, Seville, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovaculares (CIBERCV), Madrid, Spain
- Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain
| |
Collapse
|
3
|
Urocortin Role in Ischemia Cardioprotection and the Adverse Cardiac Remodeling. Int J Mol Sci 2021; 22:ijms222212115. [PMID: 34829997 PMCID: PMC8622004 DOI: 10.3390/ijms222212115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/24/2021] [Accepted: 11/05/2021] [Indexed: 11/17/2022] Open
Abstract
Despite the considerable progress in strategies of myocardial protection, ischemic heart diseases (IHD) and consequent heart failure (HF) remain the main cause of mortality worldwide. Several procedures are used routinely to guarantee the prompt and successful reestablishment of blood flow to preserve the myocardial viability of infarcted hearts from ischemia injuries. However, ischemic heart reperfusion/revascularization triggers additional damages that occur when oxygen-rich blood re-enters the vulnerable myocardial tissue, which is a phenomenon known as ischemia and reperfusion (I/R) syndrome. Complications of I/R injuries provoke the adverse cardiac remodeling, involving inflammation, mishandling of Ca2+ homeostasis, apoptotic genes activation, cardiac myocytes loss, etc., which often progress toward HF. Therefore, there is an urgent need to develop new cardioprotective therapies for IHD and HF. Compelling evidence from animal studies and pilot clinical trials in HF patients suggest that urocortin (Ucn) isoforms, which are peptides associated with stress and belonging to the corticotropin releasing factor family, have promising potential to improve cardiovascular functions by targeting many signaling pathways at different molecular levels. This review highlights the current knowledge on the role of urocortin isoforms in cardioprotection, focusing on its acute and long-term effects.
Collapse
|
4
|
Guijarro-Belmar A, Domanski DM, Bo X, Shewan D, Huang W. The therapeutic potential of targeting exchange protein directly activated by cyclic adenosine 3',5'-monophosphate (Epac) for central nervous system trauma. Neural Regen Res 2021; 16:460-469. [PMID: 32985466 PMCID: PMC7996029 DOI: 10.4103/1673-5374.293256] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Millions of people worldwide are affected by traumatic spinal cord injury, which usually results in permanent sensorimotor disability. Damage to the spinal cord leads to a series of detrimental events including ischaemia, haemorrhage and neuroinflammation, which over time result in further neural tissue loss. Eventually, at chronic stages of traumatic spinal cord injury, the formation of a glial scar, cystic cavitation and the presence of numerous inhibitory molecules act as physical and chemical barriers to axonal regrowth. This is further hindered by a lack of intrinsic regrowth ability of adult neurons in the central nervous system. The intracellular signalling molecule, cyclic adenosine 3′,5′-monophosphate (cAMP), is known to play many important roles in the central nervous system, and elevating its levels as shown to improve axonal regeneration outcomes following traumatic spinal cord injury in animal models. However, therapies directly targeting cAMP have not found their way into the clinic, as cAMP is ubiquitously present in all cell types and its manipulation may have additional deleterious effects. A downstream effector of cAMP, exchange protein directly activated by cAMP 2 (Epac2), is mainly expressed in the adult central nervous system, and its activation has been shown to mediate the positive effects of cAMP on axonal guidance and regeneration. Recently, using ex vivo modelling of traumatic spinal cord injury, Epac2 activation was found to profoundly modulate the post-lesion environment, such as decreasing the activation of astrocytes and microglia. Pilot data with Epac2 activation also suggested functional improvement assessed by in vivo models of traumatic spinal cord injury. Therefore, targeting Epac2 in traumatic spinal cord injury could represent a novel strategy in traumatic spinal cord injury repair, and future work is needed to fully establish its therapeutic potential.
Collapse
Affiliation(s)
- Alba Guijarro-Belmar
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen; Sainsbury Wellcome Centre, University College London, London, UK
| | - Dominik Mateusz Domanski
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen, UK
| | - Xuenong Bo
- Center for Neuroscience, Surgery and Trauma, Queen Mary University of London, London, UK
| | - Derryck Shewan
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen, UK
| | - Wenlong Huang
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
5
|
Lu M, Qin X, Yao J, Yang Y, Zhao M, Sun L. MiR
‐134‐5p targeting
XIAP
modulates oxidative stress and apoptosis in cardiomyocytes under hypoxia/reperfusion‐induced injury. IUBMB Life 2020; 72:2154-2166. [PMID: 32797709 DOI: 10.1002/iub.2351] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/17/2020] [Accepted: 07/03/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Min Lu
- Department of Cardiologry Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine Zhengzhou Henan China
| | - Xinglei Qin
- Department of Hepatobiliary Surgery Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine Zhengzhou Henan China
| | - Jungong Yao
- Department of Cardiologry Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine Zhengzhou Henan China
| | - Yuanyuan Yang
- Department of Cardiologry Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine Zhengzhou Henan China
| | - Minghu Zhao
- Department of Cardiologry Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine Zhengzhou Henan China
| | - Lin Sun
- Department of Cardiologry Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine Zhengzhou Henan China
| |
Collapse
|
6
|
Wang J, Liu J, Xie L, Cai X, Ma X, Gong J. Bisoprolol, a β 1 antagonist, protects myocardial cells from ischemia-reperfusion injury via PI3K/AKT/GSK3β pathway. Fundam Clin Pharmacol 2020; 34:708-720. [PMID: 32363697 DOI: 10.1111/fcp.12562] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/23/2020] [Accepted: 04/29/2020] [Indexed: 12/30/2022]
Abstract
The aim of this work was to explore whether bisoprolol plays a protective role in cardiomyocytes against ischemia-reperfusion injury via PI3K/AKT/ GSK3β pathway. We pretreated male Sprague Dawley (SD) rats with bisoprolol by oral administration prior to 0.5 h ischemia/4 h reperfusion. Myocardial infarct size and serum levels of cTnI and CK-MB were measured. In vitro, H9c2 cells were treated with hypoxia and reoxygenation, followed by measurement of cell viability, apoptosis, ROS production, cytometry, activities of AKT, GSK3β, and p-38 in the presence and absence of GSK3β siRNA. We found that bisoprolol reduced infarct size from 44% in I/R group to 31% in treated group (P < 0.05). The levels of cTnI and CK-MB were decreased from 286 ± 7 pg/mL and 32.2 ± 2 ng/mL in I/R group to 196 ± 2 pg/mL and 19.6 ± 0.9 ng/mL in the treated group, respectively (P < 0.05). Bisoprolol also increased cell viability while decreased apoptosis and ROS production in the treatment of hypoxia/ reoxygenation. Furthermore, bisoprolol increased AKT and GSK3β phosphorylation, an effect that was immediately eliminated by LY294002. GSK3β-specific siRNA experiment further confirmed that bisoprolol protected the myocardium against hypoxia/reoxygenation-induced injury via suppressing GSK3β activity. In conclusion, bisoprolol protected myocardium against ischemia-reperfusion injury via the PI3K/AKT/ GSK3β pathway.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Rd., Nanjing, 210002, Jiangsu, China
| | - Jing Liu
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Rd., Nanjing, 210002, Jiangsu, China
| | - Liang Xie
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Rd., Nanjing, 210002, Jiangsu, China
| | - Xiaomin Cai
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Rd., Nanjing, 210002, Jiangsu, China
| | - Xiaohua Ma
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Rd., Nanjing, 210002, Jiangsu, China
| | - Jianbin Gong
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Rd., Nanjing, 210002, Jiangsu, China
| |
Collapse
|
7
|
Miningou N, Blackwell KT. The road to ERK activation: Do neurons take alternate routes? Cell Signal 2020; 68:109541. [PMID: 31945453 PMCID: PMC7127974 DOI: 10.1016/j.cellsig.2020.109541] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 01/11/2020] [Accepted: 01/12/2020] [Indexed: 01/29/2023]
Abstract
The ERK cascade is a central signaling pathway that regulates a wide variety of cellular processes including proliferation, differentiation, learning and memory, development, and synaptic plasticity. A wide range of inputs travel from the membrane through different signaling pathway routes to reach activation of one set of output kinases, ERK1&2. The classical ERK activation pathway beings with growth factor activation of receptor tyrosine kinases. Numerous G-protein coupled receptors and ionotropic receptors also lead to ERK through increases in the second messengers calcium and cAMP. Though both types of pathways are present in diverse cell types, a key difference is that most stimuli to neurons, e.g. synaptic inputs, are transient, on the order of milliseconds to seconds, whereas many stimuli acting on non-neural tissue, e.g. growth factors, are longer duration. The ability to consolidate these inputs to regulate the activation of ERK in response to diverse signals raises the question of which factors influence the difference in ERK activation pathways. This review presents both experimental studies and computational models aimed at understanding the control of ERK activation and whether there are fundamental differences between neurons and other cells. Our main conclusion is that differences between cell types are quite subtle, often related to differences in expression pattern and quantity of some molecules such as Raf isoforms. In addition, the spatial location of ERK is critical, with regulation by scaffolding proteins producing differences due to colocalization of upstream molecules that may differ between neurons and other cells.
Collapse
Affiliation(s)
- Nadiatou Miningou
- Department of Chemistry and Biochemistry, George Mason University, Fairfax, VA 22030, United States of America
| | - Kim T Blackwell
- Interdisciplinary Program in Neuroscience and Bioengineering Department, George Mason University, Fairfax, VA 22030, United States of America.
| |
Collapse
|
8
|
Sun F, Yuan W, Wu H, Chen G, Sun Y, Yuan L, Zhang W, Lei M. LncRNA KCNQ1OT1 attenuates sepsis-induced myocardial injury via regulating miR-192-5p/XIAP axis. Exp Biol Med (Maywood) 2020; 245:620-630. [PMID: 32102564 DOI: 10.1177/1535370220908041] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Myocardial dysfunction is a prime cause of death in sepsis. This study is to delve into the function of lncRNA KCNQ1OT1 in myocardial injury induced by sepsis. Sepsis-induced myocardial injury model in rat was initiated by intraperitoneally injecting of LPS (10 mg/kg) in vivo, and cardiomyocyte H9c2 was treated with LPS to mimic sepsis in vitro. KCNQ1OT1 and miR-192-5p expressions were detected by qRT-PCR. The cell viability was probed with CCK-8 experiment and the apoptosis of the cardiomyocytes was tested using flow cytometry analysis. Western blot was operated to determine apoptosis-related proteins expressions. ELISA was used to evaluate the levels of TNF-α, IL-6, and IL-1β. Bioinformatics analysis, RT-PCR, dual luciferase reporter assay, and RNA immunoprecipitation experiment were utilized to detect the interrelation of genes. Herein, we proved that KCNQ1OT1 was considerably down-regulated, whereas miR-192-5p was markedly increased in myocardial tissues of septic rats. KCNQ1OT1 interrelated with miR-192-5p, and negatively modulated its expression levels. Overexpression of KCNQ1OT1 or the transfection of miR-192-5p inhibitors greatly facilitated the viability and impeded the apoptosis of H9c2 cardiomyocytes. miR-192-5p paired with the 3ʹUTR of XIAP, and repressed its protein expression, and XIAP was modulated positively by KCNQ1OT1. In conclusion, our work indicates that down-regulation of KCNQ1OT1 advances cardiac injury through regulating miR-192-5p/XIAP axis during sepsis. Impact statement Sepsis-induced cardiomyopathy remains to be a major challenge to health care systems around the globe. There are no known therapies currently available that can cure the disease. This study provides convincing evidence that KCNQ1OT1 could attenuate sepsis-mediated myocardial injury. We further demonstrate that the beneficial function of KCNQ1OT1 was achieved by regulating the miR-192-5p/XIAP axis. We therefore found a new mechanism of cardioprotective effect of KCNQ1OT1, one which also offers a novel theoretical basis for the therapy of sepsis-induced cardiomyopathy.
Collapse
Affiliation(s)
- Fangyuan Sun
- Department of Critical Care Medicine, Shanghai Seventh People's Hospital, Shanghai, 200137, China
| | - Weifang Yuan
- Department of Critical Care Medicine, Shanghai Seventh People's Hospital, Shanghai, 200137, China
| | - Hao Wu
- Trauma center, Shanghai Seventh People's Hospital, Shanghai 200137, China
| | - Gang Chen
- Department of Critical Care Medicine, Shanghai Seventh People's Hospital, Shanghai, 200137, China
| | - Yuxia Sun
- Department of Critical Care Medicine, Shanghai Seventh People's Hospital, Shanghai, 200137, China
| | - Lin Yuan
- Department of Critical Care Medicine, Shanghai Seventh People's Hospital, Shanghai, 200137, China
| | - Wei Zhang
- Trauma center, Shanghai Seventh People's Hospital, Shanghai 200137, China
| | - Ming Lei
- Department of Critical Care Medicine, Shanghai Seventh People's Hospital, Shanghai, 200137, China
| |
Collapse
|
9
|
Saul D, Geisberg LK, Gehle T, Hoffmann DB, Tezval M, Sehmisch S, Komrakova M. Changes in Musculoskeletal System and Metabolism in Osteoporotic Rats Treated With Urocortin. Front Endocrinol (Lausanne) 2019; 10:400. [PMID: 31293517 PMCID: PMC6601316 DOI: 10.3389/fendo.2019.00400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 06/06/2019] [Indexed: 01/30/2023] Open
Abstract
Objective: In aging population, postmenopausal osteoporosis and decline of musculoskeletal function, referred to as "frailty syndrome" lead to loss of bone and muscle, causing falls, and fall-related injuries. To limit the impact of this portentous duo, simultaneous treatment of both is needed. Urocortin (UCN) has been reported to improve osteoporotic bone properties while its effect on muscle has not been addressed yet. Design and Methods: We aimed to investigate the effect of urocortin in vivo on skeletal muscle structure in osteopenic rats. Sixty Sprague-Dawley rats were divided into five groups: four were ovariectomized (OVX) and one underwent sham operation (SHAM). One ovariectomized group was left untreated (OVX), while one was treated with urocortin s.c. in 3 μg/kg body weight (bw) (OVX+UCN low), one with 30 μg/kg (OVX+UCN high), while one group was treated with estradiol orally (OVX+E: 0.2 mg/kg bw), each for 35 days. Mm. gastrocnemius, longissimus, and soleus were isolated and capillary density as well as diameters of type I and II fibers were measured. In addition, we examined the effect of UCN on tibia using biomechanical, micro-CT and ashing analysis and investigated the blood serum. Results: We demonstrated a positive effect of UCN on M. soleus, in which fiber diameter was positively influenced. The biomechanical and structural parameters of bone were not changed in UCN treated rats. The higher cholesterol, glucose and triglyceride levels in the "UCN high" group raise concern about this treatment. Conclusions: Our results portray urocortin as a substance that can be assessed for future therapeutic treatments of estrogen deficiency. New and Noteworthy: Urocortin has a positive effect on M. soleus (diameter). Urocortin raises serum cholesterol and triglyceride levels. Bone tissue was not affected by UCN.
Collapse
Affiliation(s)
- Dominik Saul
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August-University of Göttingen, Göttingen, Germany
| | - Laura Katharina Geisberg
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August-University of Göttingen, Göttingen, Germany
| | - Torben Gehle
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August-University of Göttingen, Göttingen, Germany
| | - Daniel Bernd Hoffmann
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August-University of Göttingen, Göttingen, Germany
| | - Mohammad Tezval
- Klinik für Unfallchirurgie, Sporttraumatologie und Handchirurgie, Klinikum Vest, Recklinghausen, Germany
| | - Stephan Sehmisch
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August-University of Göttingen, Göttingen, Germany
| | - Marina Komrakova
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August-University of Göttingen, Göttingen, Germany
- *Correspondence: Marina Komrakova ; orcid.org/0000-0002-6225-4378
| |
Collapse
|
10
|
Liu J, Li Y, Tang Y, Cheng J, Wang J, Li J, Ma X, Zhuang W, Gong J, Liu Z. Rhein protects the myocardiac cells against hypoxia/reoxygention-induced injury by suppressing GSK3β activity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 51:1-6. [PMID: 30466606 DOI: 10.1016/j.phymed.2018.06.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 05/22/2018] [Accepted: 06/19/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Rhein, an anthraquinone compound isolated from rhubarb, has been shown to protect the pancreatic β cells from hyperglycemia induced apoptosis in our previous studies. PURPOSE In the present study, we examined whether rhein can protect myocardial cells against ischemia reperfusion (I/R)-induced apoptosis and investigated the underlying mechanism. METHODS We used an in vitro model of myocardial hypoxia/reoxygenation (H/R) injury. H9c2 cells were incubated with rhein for 1 h and then subjected to hypoxia for 6 h, followed by reoxygenation for 2 h. Cells viability, apoptosis and ROS were assayed for the treated cells. AKT, p-AKT, GSK3β, p- GSK3β, P38 and p-P38 proteins were analyzed using Western blotting. PI3K/AKT inhibitor, LY294002, and GSK3β siRNA were also used to determine the signaling pathways involved in the protection by rhein. RESULTS Rhein increased viability, decreased apoptosis and ROS production, of the cells that were exposed to H/R. Rhein also increased the phosphorylation of AKT and GSK3β, an effect that was eliminated by LY294002. GSK3β silencing by siRNA showed similar effect as LY294002. The p-P38 level was upregulated by H/R and downregulated in the presence of rhein; however, the p-P38 downregulation was completely abolished by GSK3β silencing. CONCLUSION Rhein protects myocardial H9c2 cells against hypoxia/reoxygenation induced injury via AKT/ GSK3β/p38 pathway.
Collapse
Affiliation(s)
- Jing Liu
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Yanming Li
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Yi Tang
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Jinghua Cheng
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Jing Wang
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Jianhua Li
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Xiaohua Ma
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Wei Zhuang
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Jianbin Gong
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China.
| | - Zhihong Liu
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
11
|
Grisanti LA, Schumacher SM, Tilley DG, Koch WJ. Designer Approaches for G Protein-Coupled Receptor Modulation for Cardiovascular Disease. JACC Basic Transl Sci 2018; 3:550-562. [PMID: 30175279 PMCID: PMC6115700 DOI: 10.1016/j.jacbts.2017.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 12/14/2017] [Indexed: 12/17/2022]
Abstract
The new horizon for cardiac therapy may lie beneath the surface, with the downstream mediators of G protein–coupled receptor (GPCR) activity. Targeted approaches have shown that receptor activation may be biased toward signaling through G proteins or through GPCR kinases (GRKs) and β-arrestins, with divergent functional outcomes. In addition to these canonical roles, numerous noncanonical activities of GRKs and β-arrestins have been demonstrated to modulate GPCR signaling at all levels of receptor activation and regulation. Further, research continues to identify novel GRK/effector and β-arrestin/effector complexes with distinct impacts on cardiac function in the normal heart and the diseased heart. Coupled with the identification of once orphan receptors and endogenous ligands with beneficial cardiovascular effects, this expands the repertoire of GPCR targets. Together, this research highlights the potential for focused therapeutic activation of beneficial pathways, with simultaneous exclusion or inhibition of detrimental signaling, and represents a new wave of therapeutic development.
Collapse
Key Words
- AR, adrenergic receptor
- AT1R, angiotensin II type 1A receptor
- CRF, corticotropin-releasing factor
- EGFR, epidermal growth factor receptor
- ERK1/2, extracellular signal-regulated kinase
- G protein–coupled receptor kinases
- G protein–coupled receptors
- GPCR, G protein–coupled receptor
- GRK, G protein–coupled receptor kinase
- HF, heart failure
- ICL, intracellular loop
- PI3K, phosphoinositide 3-kinase
- SERCA2a, sarco(endo)plasmic reticulum Ca2+-ATPase
- SII, [Sar(1), Ile (4), Ile(8)]-angiotensin II
- biased ligands
Collapse
Affiliation(s)
- Laurel A Grisanti
- Center for Translational Medicine and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania.,Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Sarah M Schumacher
- Center for Translational Medicine and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania.,Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Douglas G Tilley
- Center for Translational Medicine and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Walter J Koch
- Center for Translational Medicine and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
12
|
Domínguez-Rodríguez A, Mayoral-Gonzalez I, Avila-Medina J, de Rojas-de Pedro ES, Calderón-Sánchez E, Díaz I, Hmadcha A, Castellano A, Rosado JA, Benitah JP, Gomez AM, Ordoñez A, Smani T. Urocortin-2 Prevents Dysregulation of Ca 2+ Homeostasis and Improves Early Cardiac Remodeling After Ischemia and Reperfusion. Front Physiol 2018; 9:813. [PMID: 30018568 PMCID: PMC6037857 DOI: 10.3389/fphys.2018.00813] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/11/2018] [Indexed: 12/02/2022] Open
Abstract
Aims: Urocortin-2 (Ucn-2) is a potent cardioprotector against Ischemia and Reperfusion (I/R) injuries. However, little is known about its role in the regulation of intracellular Ca2+ concentration ([Ca2+]i) under I/R. Here, we examined whether the addition of Ucn-2 in reperfusion promotes cardioprotection focusing on ([Ca2+]i handling. Methods and Results: Cardiac Wistar rat model of I/R was induced by transient ligation of the left coronary artery and experiments were conducted 1 week after surgery in tissue and adult cardiomyocytes isolated from risk and remote zones. We observed that I/R promoted significant alteration in cardiac contractility as well as an increase in hypertrophy and fibrosis in both zones. The study of confocal [Ca2+]i imaging in adult cardiomyocytes revealed that I/R decreased the amplitude of [Ca2+]i transient and cardiomyocytes contraction in risk and remote zones. Interestingly, intravenous infusion of Ucn-2 before heart’s reperfusion recovered significantly cardiac contractility and prevented fibrosis, but it didn’t affect cardiac hypertrophy. Moreover, Ucn-2 recovered the amplitude of [Ca2+]i transient and modulated the expression of several proteins related to [Ca2+]i homeostasis, such as TRPC5 and Orai1 channels. Using Neonatal Rat Ventricular Myocytes (NRVM) we demonstrated that Ucn-2 blunted I/R-induced Store Operated Ca2+ Entry (SOCE), decreased the expression of TRPC5 and Orai1 as well as their interaction in reperfusion. Conclusion: Our study provides the first evidences demonstrating that Ucn-2 addition at the onset of reperfusion attenuates I/R-induced adverse cardiac remodeling, involving the [Ca2+]i handling and inhibiting the expression and interaction between TRPC5 and Orai1.
Collapse
Affiliation(s)
- Alejandro Domínguez-Rodríguez
- Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío, University of Seville, CIBERCV, CSIC, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain
| | - Isabel Mayoral-Gonzalez
- Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío, University of Seville, CIBERCV, CSIC, Seville, Spain
| | - Javier Avila-Medina
- Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío, University of Seville, CIBERCV, CSIC, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain
| | - Eva S de Rojas-de Pedro
- Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío, University of Seville, CIBERCV, CSIC, Seville, Spain
| | - Eva Calderón-Sánchez
- Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío, University of Seville, CIBERCV, CSIC, Seville, Spain
| | - Ignacio Díaz
- Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío, University of Seville, CIBERCV, CSIC, Seville, Spain
| | - Abdelkrim Hmadcha
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Junta de Andalucia, University of Pablo de Olavide, University of Seville, CSIC, Seville, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
| | - Antonio Castellano
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain
| | - Juan A Rosado
- Departamento de Fisiología, Universidad de Extremadura, Cáceres, Spain
| | - Jean-Pierre Benitah
- UMR-S 1180, INSERM, Universite Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Ana M Gomez
- UMR-S 1180, INSERM, Universite Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Antonio Ordoñez
- Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío, University of Seville, CIBERCV, CSIC, Seville, Spain
| | - Tarik Smani
- Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío, University of Seville, CIBERCV, CSIC, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
13
|
Zhang L, Zhang L, Liu H, Jiang F, Wang H, Li D, Gao R. Inhibition of Epac2 Attenuates Neural Cell Apoptosis and Improves Neurological Deficits in a Rat Model of Traumatic Brain Injury. Front Neurosci 2018; 12:263. [PMID: 29740274 PMCID: PMC5924794 DOI: 10.3389/fnins.2018.00263] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/05/2018] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of mortality and disability worldwide. TBI-induced neuronal apoptosis is one of the main contributors to the secondary injury process. The aim of this study is to investigate the involvement of Exchange protein directly activated by cAMP 2 (Epac2) on TBI. We found that the expression level of Epac2 surrounding the injured area of brain in rats of TBI model was significantly increased at 12 h after TBI. The role of Epac2 in TBI was further explored by using a selective Epac2 antagonist ESI-05 to decrease the Epac2 expression. We discovered that inhibition of Epac2 could improve the neurological impairment and attenuate brain edema following TBI. The Epac2 inhibition effectively reduced neuronal cell death and P38 MAPK signaling pathway may be involved in this process. Our results suggest that inhibition of Epac2 may be a potential therapy for TBI by reducing the neural cell death, alleviating brain edema and improving neurologic deficits.
Collapse
Affiliation(s)
- Ling Zhang
- Translational Medicine Center, The First People's Hospital of Zhangjiagang, Zhangjiagang, China.,Department of Neurosurgery, The First People's Hospital of Zhangjiagang, Zhangjiagang, China
| | - Li Zhang
- Department of Neurosurgery, The First People's Hospital of Zhangjiagang, Zhangjiagang, China
| | - Huixiang Liu
- Department of Neurosurgery, The First People's Hospital of Zhangjiagang, Zhangjiagang, China
| | - Feng Jiang
- Department of Neurosurgery, The First People's Hospital of Zhangjiagang, Zhangjiagang, China
| | - Huanjing Wang
- Department of Neurosurgery, Zhangjiagang Hospital of Traditional Chinese Medicine, Zhangjiagang, China
| | - Di Li
- Translational Medicine Center, The First People's Hospital of Zhangjiagang, Zhangjiagang, China
| | - Rong Gao
- Department of Neurosurgery, The First People's Hospital of Zhangjiagang, Zhangjiagang, China
| |
Collapse
|
14
|
Robichaux WG, Cheng X. Intracellular cAMP Sensor EPAC: Physiology, Pathophysiology, and Therapeutics Development. Physiol Rev 2018; 98:919-1053. [PMID: 29537337 PMCID: PMC6050347 DOI: 10.1152/physrev.00025.2017] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/05/2017] [Accepted: 09/06/2017] [Indexed: 12/13/2022] Open
Abstract
This review focuses on one family of the known cAMP receptors, the exchange proteins directly activated by cAMP (EPACs), also known as the cAMP-regulated guanine nucleotide exchange factors (cAMP-GEFs). Although EPAC proteins are fairly new additions to the growing list of cAMP effectors, and relatively "young" in the cAMP discovery timeline, the significance of an EPAC presence in different cell systems is extraordinary. The study of EPACs has considerably expanded the diversity and adaptive nature of cAMP signaling associated with numerous physiological and pathophysiological responses. This review comprehensively covers EPAC protein functions at the molecular, cellular, physiological, and pathophysiological levels; and in turn, the applications of employing EPAC-based biosensors as detection tools for dissecting cAMP signaling and the implications for targeting EPAC proteins for therapeutic development are also discussed.
Collapse
Affiliation(s)
- William G Robichaux
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center , Houston, Texas
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center , Houston, Texas
| |
Collapse
|
15
|
Laudette M, Zuo H, Lezoualc'h F, Schmidt M. Epac Function and cAMP Scaffolds in the Heart and Lung. J Cardiovasc Dev Dis 2018; 5:jcdd5010009. [PMID: 29401660 PMCID: PMC5872357 DOI: 10.3390/jcdd5010009] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 01/25/2018] [Accepted: 01/29/2018] [Indexed: 12/13/2022] Open
Abstract
Evidence collected over the last ten years indicates that Epac and cAMP scaffold proteins play a critical role in integrating and transducing multiple signaling pathways at the basis of cardiac and lung physiopathology. Some of the deleterious effects of Epac, such as cardiomyocyte hypertrophy and arrhythmia, initially described in vitro, have been confirmed in genetically modified mice for Epac1 and Epac2. Similar recent findings have been collected in the lung. The following sections will describe how Epac and cAMP signalosomes in different subcellular compartments may contribute to cardiac and lung diseases.
Collapse
Affiliation(s)
- Marion Laudette
- Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université Toulouse III, 31432 Toulouse, France.
| | - Haoxiao Zuo
- Department of Molecular Pharmacology, University of Groningen, 9713AV Groningen, The Netherlands.
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713AV Groningen, The Netherlands.
| | - Frank Lezoualc'h
- Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université Toulouse III, 31432 Toulouse, France.
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, 9713AV Groningen, The Netherlands.
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713AV Groningen, The Netherlands.
| |
Collapse
|
16
|
Rademaker MT, Richards AM. Urocortins: Actions in health and heart failure. Clin Chim Acta 2017; 474:76-87. [DOI: 10.1016/j.cca.2017.09.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 09/04/2017] [Indexed: 01/21/2023]
|
17
|
miR-125a, miR-139 and miR-324 contribute to Urocortin protection against myocardial ischemia-reperfusion injury. Sci Rep 2017; 7:8898. [PMID: 28827743 PMCID: PMC5566224 DOI: 10.1038/s41598-017-09198-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/21/2017] [Indexed: 12/20/2022] Open
Abstract
Urocortin 1 and 2 (Ucn-1 and Ucn-2) have established protective actions against myocardial ischemia-reperfusion (I/R) injuries. However, little is known about their role in posttranscriptional regulation in the process of cardioprotection. Herein, we investigated whether microRNAs play a role in urocortin-induced cardioprotection. Administration of Ucn-1 and Ucn-2 at the beginning of reperfusion significantly restored cardiac function, as evidenced ex vivo in Langendorff-perfused rat hearts and in vivo in rat subjected to I/R. Experiments using microarray and qRT-PCR determined that the addition of Ucn-1 at reperfusion modulated the expression of several miRNAs with unknown role in cardiac protection. Ucn-1 enhanced the expression of miR-125a-3p, miR-324-3p; meanwhile it decreased miR-139-3p. Similarly, intravenous infusion of Ucn-2 in rat model of I/R mimicked the effect of Ucn-1 on miR-324-3p and miR-139-3p. The effect of Ucn-1 involves the activation of corticotropin-releasing factor receptor-2, Epac2 and ERK1/2. Moreover, the overexpression of miR-125a-3p, miR-324-3p and miR-139-3p promoted dysregulation of genes expression involved in cell death and apoptosis (BRCA1, BIM, STAT2), in cAMP and Ca2+ signaling (PDE4a, CASQ1), in cell stress (NFAT5, XBP1, MAP3K12) and in metabolism (CPT2, FoxO1, MTRF1, TAZ). Altogether, these data unveil a novel role of urocortin in myocardial protection, involving posttranscriptional regulation with miRNAs.
Collapse
|
18
|
Wu D, Wang J, Wang H, Ji A, Li Y. Protective roles of bioactive peptides during ischemia-reperfusion injury: From bench to bedside. Life Sci 2017; 180:83-92. [PMID: 28527782 DOI: 10.1016/j.lfs.2017.05.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 12/14/2022]
Abstract
Ischemia-reperfusion (I/R) is a well-known pathological condition which may lead to disability and mortality. I/R injury remains an unresolved and complicated situation in a number of clinical conditions, such as cardiac arrest with successful reanimation, as well as ischemic events in brain and heart. Peptides have many attractive advantages which make them suitable candidate drugs in treating I/R injury, such as low toxicity and immunogenicity, good solubility property, distinct tissue distribution pattern, and favorable pharmacokinetic profile. An increasing number of studies indicate that peptides could protect against I/R injury in many different organs and tissues. Peptides also face several therapeutic challenges that limit their clinical application. In this review, we present the mechanisms of action of peptides in reducing I/R injury, as well as further discuss modification strategies to improve the functional properties of bioactive peptides.
Collapse
Affiliation(s)
- Dongdong Wu
- Henan University School of Basic Medical Sciences, Kaifeng 475004, Henan, China; Institute of Environmental Medicine, Henan University, Kaifeng 475004, Henan, China
| | - Jun Wang
- Henan University School of Basic Medical Sciences, Kaifeng 475004, Henan, China; Institute of Environmental Medicine, Henan University, Kaifeng 475004, Henan, China
| | - Honggang Wang
- Henan University School of Basic Medical Sciences, Kaifeng 475004, Henan, China; Institute of Environmental Medicine, Henan University, Kaifeng 475004, Henan, China
| | - Ailing Ji
- Henan University School of Basic Medical Sciences, Kaifeng 475004, Henan, China; Institute of Environmental Medicine, Henan University, Kaifeng 475004, Henan, China.
| | - Yanzhang Li
- Henan University School of Basic Medical Sciences, Kaifeng 475004, Henan, China; Institute of Environmental Medicine, Henan University, Kaifeng 475004, Henan, China.
| |
Collapse
|
19
|
Khaliulin I, Bond M, James AF, Dyar Z, Amini R, Johnson JL, Suleiman MS. Functional and cardioprotective effects of simultaneous and individual activation of protein kinase A and Epac. Br J Pharmacol 2017; 174:438-453. [PMID: 28071786 PMCID: PMC5323515 DOI: 10.1111/bph.13709] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 12/23/2016] [Accepted: 01/05/2017] [Indexed: 01/16/2023] Open
Abstract
Background and Purpose Myocardial cAMP elevation confers cardioprotection against ischaemia/reperfusion (I/R) injury. cAMP activates two independent signalling pathways, PKA and Epac. This study investigated the cardiac effects of activating PKA and/or Epac and their involvement in cardioprotection against I/R. Experimental Approach Hearts from male rats were used either for determination of PKA and PKC activation or perfused in the Langendorff mode for either cardiomyocyte isolation or used to monitor functional activity at basal levels and after 30 min global ischaemia and 2 h reperfusion. Functional recovery and myocardial injury during reperfusion (LDH release and infarct size) were evaluated. Activation of PKA and/or Epac in perfused hearts was induced using cell permeable cAMP analogues in the presence or absence of inhibitors of PKA, Epac and PKC. H9C2 cells and cardiomyocytes were used to assess activation of Epac and effect on Ca2+ transients. Key Results Selective activation of either PKA or Epac was found to trigger a positive inotropic effect, which was considerably enhanced when both pathways were simultaneously activated. Only combined activation of PKA and Epac induced marked cardioprotection against I/R injury. This was accompanied by PKCε activation and repressed by inhibitors of PKA, Epac or PKC. Conclusion and Implications Simultaneous activation of both PKA and Epac induces an additive inotropic effect and confers optimal and marked cardioprotection against I/R injury. The latter effect is mediated by PKCε activation. This work has introduced a new therapeutic approach and targets to protect the heart against cardiac insults.
Collapse
Affiliation(s)
- Igor Khaliulin
- School of Clinical Sciences and Bristol Cardiovascular, University of Bristol, Bristol, UK
| | - Mark Bond
- School of Clinical Sciences and Bristol Cardiovascular, University of Bristol, Bristol, UK
| | - Andrew F James
- School of Clinical Sciences and Bristol Cardiovascular, University of Bristol, Bristol, UK
| | - Zara Dyar
- School of Clinical Sciences and Bristol Cardiovascular, University of Bristol, Bristol, UK
| | - Raheleh Amini
- School of Clinical Sciences and Bristol Cardiovascular, University of Bristol, Bristol, UK
| | - Jason L Johnson
- School of Clinical Sciences and Bristol Cardiovascular, University of Bristol, Bristol, UK
| | - M-Saadeh Suleiman
- School of Clinical Sciences and Bristol Cardiovascular, University of Bristol, Bristol, UK
| |
Collapse
|
20
|
Li N, Shang L, Wang SC, Liao LS, Chen D, Huang JF, Xiong K. The Toxic Effect of ALLN on Primary Rat Retinal Neurons. Neurotox Res 2016; 30:392-406. [PMID: 27194525 DOI: 10.1007/s12640-016-9624-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 04/04/2016] [Accepted: 04/16/2016] [Indexed: 10/21/2022]
Abstract
N-acetyl-leucyl-leucyl-norleucinal (ALLN), an inhibitor of proteasomes and calpain, is widely used to reduce proteasomes or calpain-mediated cell death in rodents. However, ALLN is toxic to retinal neurons to some extent. At the concentration of 10 μM, ALLN is non-toxic to cortical neurons, but induces cell death of retinal neurons in vitro. The tolerance concentration of ALLN for retinal neurons is unclear, and the precise mechanism of cell death induced by ALLN remains elusive. In this study, we investigated the toxic effect of ALLN on primary retinal neurons. The 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay showed no significant changes of cell viability at 1 μM but decreased cell viability after treatment of ALLN at 2.5, 5, and 7.5 μM. Lactate dehydrogenase (LDH) release was highly elevated and propidium iodide (PI)-positive cells were significantly increased at 2.5, 5, and 7.5 μM after all treatment times. Moreover, the protein levels of caspase-3 were up-regulated at 5 and 7.5 μM after 12 and 24 h of ALLN treatment. The ratio of Bax/Bcl-2 was raised and Annexin V-positive cells were increased at 5 and 7.5 μM after 12 and 24 h of ALLN treatment. However, there were no significant changes in either the ratio of microtubule-associated protein 1 light chain 3 (LC3) II/LC3 I or monodansylcadaverine (MDC) staining. Our data clearly show that at the concentrations equal to and higher than 2.5 μM, ALLN may induce cell death of primary retinal neurons by necrosis and apoptosis, but not autophagy. These suggest that primary retinal neurons are more susceptible to ALLN treatment and provide a possible mechanism for the cell death of ALLN-sensitive cells in ALLN injury.
Collapse
Affiliation(s)
- Na Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Lei Shang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Shu-Chao Wang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Lv-Shuang Liao
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Dan Chen
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Ju-Fang Huang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China.
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China.
| |
Collapse
|