1
|
Millan I, Pérez S, Rius-Pérez S, Asensi MÁ, Vento M, García-Verdugo JM, Torres-Cuevas I. Postnatal hypoxic preconditioning attenuates lung damage from hyperoxia in newborn mice. Pediatr Res 2024:10.1038/s41390-024-03457-0. [PMID: 39317699 DOI: 10.1038/s41390-024-03457-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND Preterm infants frequently require oxygen supplementation at birth. However, preterm lung is especially sensible to structural and functional damage caused by oxygen free radicals. METHODS The adaptive mechanisms implied in the fetal-neonatal transition from a lower to a higher oxygen environment were evaluated in a murine model using a custom-designed oxy-chamber. Pregnant mice were randomly assigned to deliver in 14% (hypoxic preconditioning group) or 21% (normoxic group) oxygen environment. Eight hours after birth FiO2 was increased to 100% for 60 min and then switched to 21% in both groups. A control group remained in 21% oxygen throughout the study. RESULTS Mice in the normoxic group exhibited thinning of the alveolar septa, increased cell death, increased vascular damage, and decreased synthesis of pulmonary surfactant. However, lung histology, lamellar bodies microstructure, and surfactant integrity were preserved in the hypoxic preconditioning group after the hyperoxic insult. CONCLUSION Postnatal hyperoxia has detrimental effects on lung structure and function when preceded by normoxia compared to controls. However, postnatal hypoxic preconditioning mitigates lung damage caused by a hyperoxic insult. IMPACT Hypoxic preconditioning, implemented shortly after birth mitigates lung damage caused by postnatal supplemental oxygenation. The study introduces an experimental mice model to investigate the effects of hypoxic preconditioning and its effects on lung development. This model enables researchers to delve into the intricate processes involved in postnatal lung maturation. Our findings suggest that hypoxic preconditioning may reduce lung parenchymal damage and increase pulmonary surfactant synthesis in reoxygenation strategies during postnatal care.
Collapse
Affiliation(s)
- Iván Millan
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), Valencia, Spain
- Laboratory of Comparative Neurobiology, Instituto Cavanilles de Biodiversidad y Biologia Evolutiva, University of Valencia, Paterna, Valencia, Spain
| | - Salvador Pérez
- Department of Physiology, University of Valencia, Burjassot, Spain
| | - Sergio Rius-Pérez
- Department of Cell Biology, Functional Biology and Physical Anthropology, University of Valencia, Burjassot, Spain
| | | | - Máximo Vento
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), Valencia, Spain.
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), Valencia, Spain.
| | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Instituto Cavanilles de Biodiversidad y Biologia Evolutiva, University of Valencia, Paterna, Valencia, Spain
| | - Isabel Torres-Cuevas
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), Valencia, Spain.
- Department of Physiology, University of Valencia, Burjassot, Spain.
| |
Collapse
|
2
|
Alekseeva TM, Topuzova MP, Kulikov VP, Kovzelev PD, Kosenko MG, Tregub PP. Hypercapnic hypoxia as a rehabilitation method for patients after ischemic stroke. Neurol Res 2024; 46:695-705. [PMID: 38643375 DOI: 10.1080/01616412.2024.2343510] [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: 01/24/2024] [Accepted: 04/03/2024] [Indexed: 04/22/2024]
Abstract
INTRODUCTION Experimental studies on animals have demonstrated a higher neuroprotective efficacy of hypercapnic hypoxia compared to normocapnic hypoxia. Respiratory training with hypercapnic hypoxia has shown a positive impact on the functional state of the nervous system in children with cerebral palsy (CP). It can be presumed that the combined effect of moderate hypercapnia and hypoxia will be promising for clinical application within the context of early rehabilitation after ischemic stroke. METHODS A randomized triple-blind placebo-controlled study was conducted on 102 patients with ischemic stroke, aged 63.07 ± 12.1 years. All patients were diagnosed with ischemic stroke based on neuroimaging criteria and/or clinical criteria within the 48-72 hour timeframe. The experimental group (n = 50) underwent daily respiratory training with hypercapnic hypoxia (FetCO2 5-6%, FetO2 15-16%) using the 'Carbonic' device for 7-11 sessions of 20 minutes each day during the treatment process. The control group (placebo, n = 52) underwent training on a similar device modified for breathing atmospheric air. Neurological examinations were conducted on all patients before the study and on the day after completing the training course. RESULTS The standard treatment demonstrated effectiveness in terms of neurological status scales in both groups. Intermittent exposure to hypercapnic hypoxia proved more effective in improving neurological function indicators in patients compared to the placebo group: NIHSS scale scores were 40% lower than in the placebo group (p < 0.001); mRS scale scores were 35% lower (p < 0.001); B-ADL-I and RMI indices were higher by 26% (p < 0.01) and 36% (p < 0.001), respectively; MoCA scale results were 13% higher (p < 0.05); HADS and BDI-II scale scores were lower by 35% (p < 0.05) and 25% (p < 0.05), respectively. The increase in MMSE scale scores in the intervention group was 54% higher (p < 0.001), and MoCA scale scores increased by 25% (p < 0.001). CONCLUSION Respiratory training with hypercapnic hypoxia improves the functional state of the nervous system in patients with ischemic stroke. After conducting further clarifying studies, hypercapnic hypoxia can be considered as an effective method of neurorehabilitation, which can be used as early as 48-72 hours after the onset of stroke.
Collapse
Affiliation(s)
- Tatiana M Alekseeva
- Department of Neurology with Clinic, V.A. Almazov National Medical Research Center, Saint-Petersburg, Russia
| | - Maria P Topuzova
- Department of Neurology with Clinic, V.A. Almazov National Medical Research Center, Saint-Petersburg, Russia
| | - Vladimir P Kulikov
- Department of Ultrasound and Functional Diagnostics, Altai State Medical University, Barnaul, Russia
| | - Pavel D Kovzelev
- Department of Neurology with Clinic, Smart Clinic ltd, Saint-Petersburg, Russia
| | - Mark G Kosenko
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Pavel P Tregub
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
- Scientific and Educational Resource Center "Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis", RUDN University, Moscow, Russia
| |
Collapse
|
3
|
Nuszkiewicz J, Kukulska-Pawluczuk B, Piec K, Jarek DJ, Motolko K, Szewczyk-Golec K, Woźniak A. Intersecting Pathways: The Role of Metabolic Dysregulation, Gastrointestinal Microbiome, and Inflammation in Acute Ischemic Stroke Pathogenesis and Outcomes. J Clin Med 2024; 13:4258. [PMID: 39064298 PMCID: PMC11278353 DOI: 10.3390/jcm13144258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/13/2024] [Accepted: 07/20/2024] [Indexed: 07/28/2024] Open
Abstract
Acute ischemic stroke (AIS) remains a major cause of mortality and long-term disability worldwide, driven by complex and multifaceted etiological factors. Metabolic dysregulation, gastrointestinal microbiome alterations, and systemic inflammation are emerging as significant contributors to AIS pathogenesis. This review addresses the critical need to understand how these factors interact to influence AIS risk and outcomes. We aim to elucidate the roles of dysregulated adipokines in obesity, the impact of gut microbiota disruptions, and the neuroinflammatory cascade initiated by lipopolysaccharides (LPS) in AIS. Dysregulated adipokines in obesity exacerbate inflammatory responses, increasing AIS risk and severity. Disruptions in the gut microbiota and subsequent LPS-induced neuroinflammation further link systemic inflammation to AIS. Advances in neuroimaging and biomarker development have improved diagnostic precision. Here, we highlight the need for a multifaceted approach to AIS management, integrating metabolic, microbiota, and inflammatory insights. Potential therapeutic strategies targeting these pathways could significantly improve AIS prevention and treatment. Future research should focus on further elucidating these pathways and developing targeted interventions to mitigate the impacts of metabolic dysregulation, microbiome imbalances, and inflammation on AIS.
Collapse
Affiliation(s)
- Jarosław Nuszkiewicz
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| | - Beata Kukulska-Pawluczuk
- Department of Neurology, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 M. Skłodowskiej—Curie St., 85-094 Bydgoszcz, Poland; (B.K.-P.); (K.P.)
| | - Katarzyna Piec
- Department of Neurology, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 M. Skłodowskiej—Curie St., 85-094 Bydgoszcz, Poland; (B.K.-P.); (K.P.)
| | - Dorian Julian Jarek
- Student Research Club of Medical Biology and Biochemistry, Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| | - Karina Motolko
- Student Research Club of Neurology, Department of Neurology, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 M. Skłodowskiej—Curie St., 85-094 Bydgoszcz, Poland;
| | - Karolina Szewczyk-Golec
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| |
Collapse
|
4
|
Gheitasi I, Akbari G, Savari F. Physiological and cellular mechanisms of ischemic preconditioning microRNAs-mediated in underlying of ischemia/reperfusion injury in different organs. Mol Cell Biochem 2024:10.1007/s11010-024-05052-7. [PMID: 39001984 DOI: 10.1007/s11010-024-05052-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/10/2024] [Indexed: 07/15/2024]
Abstract
Ischemia-reperfusion (I/R) injury, as a pathological phenomenon, takes place when blood supply to an organ is disrupted and then aggravated during restoration of blood flow. Ischemic preconditioning (IPC) is a potent method for attenuating subsequent events of IR damage in numerous organs. IPC protocol is determined by a brief and sequential time periods of I/R before the main ischemia. MicroRNAs are endogenous non-coding RNAs that regulate post-transcriptionally target mRNA translation via degrading it and/or suppressing protein synthesis. This review introduces the physiological and cellular mechanisms of ischemic preconditioning microRNAs-mediated after I/R insult in different organs such as the liver, kidney, heart, brain, and intestine. Data of this review have been collected from the scientific articles published in databases such as Science Direct, Scopus, PubMed, Web of Science, and Scientific Information Database from 2000 to 2023. Based on these literature studies, IPC/IR intervention can affect cellular mechanisms including oxidative stress, apoptosis, angiogenesis, and inflammation through up-regulation or down-regulation of multiple microRNAs and their target genes.
Collapse
Affiliation(s)
- Izadpanah Gheitasi
- Department of Physiology, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Ghaidafeh Akbari
- Department of Physiology, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Feryal Savari
- Department of Medical Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran.
| |
Collapse
|
5
|
Liu ZY, Lin LC, Liu ZY, Yang JJ, Tao H. m6A epitranscriptomic and epigenetic crosstalk in cardiac fibrosis. Mol Ther 2024; 32:878-889. [PMID: 38311850 PMCID: PMC11163196 DOI: 10.1016/j.ymthe.2024.01.037] [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: 11/16/2023] [Revised: 12/27/2023] [Accepted: 01/31/2024] [Indexed: 02/06/2024] Open
Abstract
Cardiac fibrosis, a crucial pathological characteristic of various cardiac diseases, presents a significant treatment challenge. It involves the deposition of the extracellular matrix (ECM) and is influenced by genetic and epigenetic factors. Prior investigations have predominantly centered on delineating the substantial influence of epigenetic and epitranscriptomic mechanisms in driving the progression of fibrosis. Recent studies have illuminated additional avenues for modulating the progression of fibrosis, offering potential solutions to the challenging issues surrounding fibrosis treatment. In the context of cardiac fibrosis, an intricate interplay exists between m6A epitranscriptomic and epigenetics. This interplay governs various pathophysiological processes: mitochondrial dysfunction, mitochondrial fission, oxidative stress, autophagy, apoptosis, pyroptosis, ferroptosis, cell fate switching, and cell differentiation, all of which affect the advancement of cardiac fibrosis. In this comprehensive review, we meticulously analyze pertinent studies, emphasizing the interplay between m6A epitranscriptomics and partial epigenetics (including histone modifications and noncoding RNA), aiming to provide novel insights for cardiac fibrosis treatment.
Collapse
Affiliation(s)
- Zhi-Yan Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China
| | - Li-Chan Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China
| | - Zhen-Yu Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China.
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China; Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China; Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China.
| |
Collapse
|
6
|
Lei L, Liu M, Ma D, Lei X, Zeng S, Li P, Huang K, Lyu J, Lei Q. Cardioprotective effects of high-altitude adaptation in cardiac surgical patients: a retrospective cohort study with propensity score matching. Front Cardiovasc Med 2024; 11:1347552. [PMID: 38628317 PMCID: PMC11019029 DOI: 10.3389/fcvm.2024.1347552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
Background The cardioprotective effect of remote ischemia preconditioning in clinical studies is inconsistent with experimental results. Adaptation to high-altitude hypoxia has been reported to be cardioprotective in animal experiments. However, the clinical significance of the cardioprotective effect of high-altitude adaptation has not been demonstrated. Methods A retrospective cohort study with propensity score matching was designed to compare the outcomes of cardiac surgery between highlanders and lowlanders in a tertiary teaching hospital. The data of adult cardiac surgical patients from January 2013 to December 2022, were collected for analysis. Patients with cardiopulmonary bypass and cardioplegia were divided into a low-altitude group (<1,500 m) and a high-altitude group (≥1,500 m) based on the altitude of their place of residence. Results Of 3,020 patients, the majority (87.5%) permanently lived in low-altitude regions [495 (435, 688) m], and there were 379 patients (12.5%) in the high-altitude group [2,552 (1,862, 3,478) m]. The 377 highlander patients were matched with lowlander patients at a ratio of 1:1. The high-altitude group exhibited a 44.5% reduction in the incidence of major adverse cardiovascular events (MACEs) compared with the low-altitude group (6.6% vs. 11.9%, P = 0.017). The patients in the moderate high-altitude subgroup (2,500-3,500 m) had the lowest incidence (5.6%) of MACEs among the subgroups. The level of creatinine kinase muscle-brain isoenzymes on the first postoperative morning was lower in the high-altitude group than in the low-altitude group (66.5 [47.9, 89.0] U/L vs. 69.5 [49.3, 96.8] U/L, P = 0.003). Conclusions High-altitude adaptation exhibits clinically significant cardioprotection in cardiac surgical patients.
Collapse
Affiliation(s)
- Li Lei
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Mengxue Liu
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Die Ma
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xia Lei
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Si Zeng
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Peng Li
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Keli Huang
- Department of Cardiac Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Juanjuan Lyu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Qian Lei
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
7
|
Pugazenthi S, Norris AJ, Lauzier DC, Lele AV, Huguenard A, Dhar R, Zipfel GJ, Athiraman U. Conditioning-based therapeutics for aneurysmal subarachnoid hemorrhage - A critical review. J Cereb Blood Flow Metab 2024; 44:317-332. [PMID: 38017387 PMCID: PMC10870969 DOI: 10.1177/0271678x231218908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/08/2023] [Accepted: 11/19/2023] [Indexed: 11/30/2023]
Abstract
Aneurysmal subarachnoid hemorrhage (SAH) carries significant mortality and morbidity, with nearly half of SAH survivors having major cognitive dysfunction that impairs their functional status, emotional health, and quality of life. Apart from the initial hemorrhage severity, secondary brain injury due to early brain injury and delayed cerebral ischemia plays a leading role in patient outcome after SAH. While many strategies to combat secondary brain injury have been developed in preclinical studies and tested in late phase clinical trials, only one (nimodipine) has proven efficacious for improving long-term functional outcome. The causes of these failures are likely multitude, but include use of therapies targeting only one element of what has proven to be multifactorial brain injury process. Conditioning is a therapeutic strategy that leverages endogenous protective mechanisms to exert powerful and remarkably pleiotropic protective effects against injury to all major cell types of the CNS. The aim of this article is to review the current body of evidence for the use of conditioning agents in SAH, summarize the underlying neuroprotective mechanisms, and identify gaps in the current literature to guide future investigation with the long-term goal of identifying a conditioning-based therapeutic that significantly improves functional and cognitive outcomes for SAH patients.
Collapse
Affiliation(s)
- Sangami Pugazenthi
- Department of Neurological Surgery, Washington University, St. Louis MO, USA
| | - Aaron J Norris
- Department of Anesthesiology, Washington University, St. Louis MO, USA
| | - David C Lauzier
- Department of Neurological Surgery, University of California, Los Angeles, CA, USA
| | - Abhijit V Lele
- Department of Anesthesiology, University of Washington, Seattle, WA, USA
| | - Anna Huguenard
- Department of Neurological Surgery, Washington University, St. Louis MO, USA
| | - Rajat Dhar
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - Gregory J Zipfel
- Departments of Neurological Surgery and Neurology, Washington University, St. Louis, MO, USA
| | - Umeshkumar Athiraman
- Department of Anesthesiology and Neurological Surgery, Washington University, St. Louis, MO, USA
| |
Collapse
|
8
|
Yoshihara T. [Imaging of In Vivo Oxygen Tension Based on Phosphorescence Lifetime Microscopy]. YAKUGAKU ZASSHI 2024; 144:275-283. [PMID: 38432937 DOI: 10.1248/yakushi.23-00168-1] [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] [Indexed: 03/05/2024]
Abstract
Molecular oxygen plays essential roles in aerobic organisms as a terminal electron acceptor in the electron transport chain in mitochondria. The intracellular oxygen concentration of the entire body is strictly regulated by a balance between the supply of oxygen from blood vessels and the consumption of oxygen in mitochondria. The disruption of oxygen homeostasis in the body often results in serious pathologies such as cancer, cerebral infarction, and chronic kidney disease, and thus considerable effort has been devoted to the development of suitable techniques allowing the qualitative and quantitative detection of tissue oxygen levels. This review focuses on recent advances in the visualization of oxygen levels in tissue based on phosphorescence lifetime measurements using exogenously small molecular oxygen probes. Specially, I introduce the principle of oxygen sensing by means of phosphorescence quenching, recent advances in intracellular and intravascular oxygen probes based on iridium(III) complexes, a system for measuring phosphorescence lifetime combined with confocal scanning microscopy, and the applications of these technologies to in vivo oxygen measurements, emphasizing the usefulness of iridium(III) complexes as biological oxygen probes.
Collapse
|
9
|
Zhang YN, Wu Q, Zhang NN, Chen HS. Ischemic Preconditioning Alleviates Cerebral Ischemia-Reperfusion Injury by Interfering With Glycocalyx. Transl Stroke Res 2023; 14:929-940. [PMID: 36168082 DOI: 10.1007/s12975-022-01081-w] [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: 06/23/2022] [Revised: 08/08/2022] [Accepted: 09/12/2022] [Indexed: 10/14/2022]
Abstract
Ischemic preconditioning (IPC) could protect the blood-brain barrier (BBB), but the underlying mechanism is not well understood. This preclinical study aimed to investigate whether glycocalyx could be involved in the neuroprotective effect of IPC on cerebral ischemia-reperfusion injury (IRI) and the possible mechanism in rat middle cerebral artery occlusion/reperfusion (MCAO/R) model. Neurological deficit scores, infarct volume, and brain edema were measured to assess the neuroprotection of IPC. Several serum biomarkers related to glycocalyx damage, such as hyaluronic acid (HA), heparan sulfate (HS), and syndecan-1 (SYND1), were evaluated, and their changes were normalized to the ratio of postoperative/preoperative concentration. Western blot and immunofluorescence were used to evaluate the content and cellular location of HA-related metabolic enzymes. This study found that (1) IPC improved brain infarction and edema, neurological impairment, and BBB disruption in IRI rats; (2) IPC significantly up-regulated HA ratio and down-regulated HS ratio, but did not affect SYND1 ratio compared with the IRI group. Moreover, the increased HA ratio was negatively related to brain edema and neurological deficit score. (3) IPC affected HA metabolism by up-regulating hyaluronate synthase-1 and matrix metalloproteinase-2, and down-regulating hyaluronidase-1 in brain tissue. Together, this is the first report that the neuroprotective effect of IPC on IRI may be mediated through interfering with glycocalyx in the MCAO/R model.
Collapse
Affiliation(s)
- Yi-Na Zhang
- Department of Neurology, General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, 110016, China
- Department of Neurology, General Hospital of Northern Theater Command of China Medical University, Shenyang, 110013, China
| | - Qiong Wu
- Department of Neurology, General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, 110016, China
| | - Nan-Nan Zhang
- Department of Neurology, General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, 110016, China
| | - Hui-Sheng Chen
- Department of Neurology, General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, 110016, China.
| |
Collapse
|
10
|
Baranova K, Nalivaeva N, Rybnikova E. Neuroadaptive Biochemical Mechanisms of Remote Ischemic Conditioning. Int J Mol Sci 2023; 24:17032. [PMID: 38069355 PMCID: PMC10707673 DOI: 10.3390/ijms242317032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
This review summarizes the currently known biochemical neuroadaptive mechanisms of remote ischemic conditioning. In particular, it focuses on the significance of the pro-adaptive effects of remote ischemic conditioning which allow for the prevention of the neurological and cognitive impairments associated with hippocampal dysregulation after brain damage. The neuroimmunohumoral pathway transmitting a conditioning stimulus, as well as the molecular basis of the early and delayed phases of neuroprotection, including anti-apoptotic, anti-oxidant, and anti-inflammatory components, are also outlined. Based on the close interplay between the effects of ischemia, especially those mediated by interaction of hypoxia-inducible factors (HIFs) and steroid hormones, the involvement of the hypothalamic-pituitary-adrenocortical system in remote ischemic conditioning is also discussed.
Collapse
Affiliation(s)
| | | | - Elena Rybnikova
- I. P. Pavlov Institute of Physiology, Russian Academy of Sciences, 199034 Saint Petersburg, Russia; (K.B.); (N.N.)
| |
Collapse
|
11
|
Sharafkhaneh A, Agrawal R, Nambi V, BaHammam A, Razjouyan J. Obesity paradox or hypoxia preconditioning: How obstructive sleep apnea modifies the Obesity-MI relationship. Sleep Med 2023; 110:132-136. [PMID: 37574613 PMCID: PMC10529841 DOI: 10.1016/j.sleep.2023.07.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/17/2023] [Accepted: 07/27/2023] [Indexed: 08/15/2023]
Abstract
OBJECTIVES The objective of this study was to evaluate the interaction between obesity and obstructive sleep apnea on acute MI in hospital mortality. METHODS This retrospective cohort study utilized Veterans Health Administration data from years 1999-2020. Participants were categorized according to their body mass index (BMI) to non-obese (BMI <30) and obese (BMI ≥30) groups. Clinical obstructive sleep apnea (SA) diagnosis was confirmed using ICD9/10 codes and the study subgroups included non-obese with no obstructive sleep apnea (nOB-nSA), non-Obese with obstructive sleep apnea (nOB-SA), obese with no obstructive sleep apnea (OB-nSA), and obese with obstructive sleep apnea (OB-SA). The primary outcome was odds ratio of in-hospital mortality during the hospitalization with acute MI as the principal diagnosis adjusted for age, gender, race, ethnicity, and Charlson comorbidity index (CCI) with the nOB-nSA group as the comparison group. RESULTS Among 72,036 veterans with acute-MI hospitalization, individuals with obesity and obstructive sleep apnea (OB-SA) had the lowest in-hospital mortality rate (1.0%) compared to those without obesity and obstructive sleep apnea (nOB-nSA, 2.8%), with obesity but without obstructive sleep apnea (OB-nSA, 2.4%), and with obesity and obstructive sleep apnea (nOB-SA, 1.4%). The adjusted odds ratio for mortality, compared to nOB-nSA, was 9% higher but not significant in OB-nSA (aOR, 1.09, 95%CI: 0.95, 1.25), 46% lower in OB-nSA (aOR, 0.54, 95%CI: 0.45, 0.66), and 52% lower in OB-SA (aOR, 0.48: 95%CI: 0.41, 0.57). CONCLUSION Our data suggest that the association between obesity and improved survival in acute MI is largely driven by the presence of sleep apnea.
Collapse
Affiliation(s)
- Amir Sharafkhaneh
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Pulmonary, Critical Care and Sleep Medicine Section, Medical Care Line, Michael E. DeBakey VA Medical Center, Houston, TX, USA.
| | - Ritwick Agrawal
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Pulmonary, Critical Care and Sleep Medicine Section, Medical Care Line, Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Vijay Nambi
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Cardiology Section, Medical Care Line, Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Ahmed BaHammam
- Department of Medicine, University Sleep Disorders Center and Pulmonary Service, King Saud University, Riyadh, Saudi Arabia; Strategic Technologies Program of the National Plan for Sciences and Technology and Innovation in the Kingdom of Saudi Arabia, Saudi Arabia
| | - Javad Razjouyan
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Center for Innovations in Quality, Effectiveness, and Safety, Michael E. DeBakey VA Medical Center, Houston, TX, USA; South Central Mental Illness Research, Education, and Clinical Center, Houston, TX, USA; Big Data Scientist Training Enhancement Program (BD-STEP), VA Office of Research and Development, Washington, DC, USA
| |
Collapse
|
12
|
Shi W, Ren C, Zhang W, Gao C, Yu W, Ji X, Chang L. Hypoxic Postconditioning Promotes Angiogenesis After Ischemic Stroke. Neuroscience 2023; 526:35-47. [PMID: 37331689 DOI: 10.1016/j.neuroscience.2023.06.009] [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: 02/12/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 06/20/2023]
Abstract
Although hypoxic postconditioning (HPC) has a protective effect on ischemic stroke, its effect on angiogenesis after ischemic stroke is still unclear. This study was designed to investigate the effects of HPC on angiogenesis after ischemic stroke and to preliminarily study the mechanism involved. Oxygen-glucose deprivation (OGD)-intervened bEnd.3 (mouse brain-derived Endothelial cell. 3) was used to simulate cerebral ischemia. Cell counting kit-8 (CCK-8), Cell BrdU proliferation, wound healing, Transwell and tube formation assays were used to evaluate the effect of HPC on the cell viability, proliferation, migration (horizontal and vertical migration), morphogenesis and tube formation of bEnd.3. A middle cerebral artery occlusion (MCAO) model was made in C57 mice to simulate focal cerebral ischemia. Rod rotation test, corner test, modified neurological severity score (mNSS), and balance beam walking test were used to evaluate the effect of HPC on the neurological impairment of mice. Immunofluorescence staining was used to evaluate the effect of HPC on angiogenesis in mice. The angiogenesis-related proteins were evaluated and quantified using western blot. Results showed that HPC significantly promoted proliferation, migration and tube formation of bEnd.3. HPC significantly reversed the neurological deficit of MCAO mice. Moreover, HPC significantly promoted angiogenesis in the peri-infarct area, and angiogenesis was found to be positively correlated with the improvement of neurological impairment. The HPC mice showed higher PLCλ and ALK5 than did MCAO. We conclude that HPC improves the neurological deficit caused by focal cerebral ischemia by promoting angiogenesis. Furthermore, the effect of HPC on improving angiogenesis may be related to PLCλ and ALK5.
Collapse
Affiliation(s)
- Wenjie Shi
- North China University of Science and Technology Affiliated Hospital, Tangshan 063000, China; Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Center of Stroke, Beijing Institute for Brain Disorder, Capital Medical University, Beijing 100053, China
| | - Wei Zhang
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Chen Gao
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Wantong Yu
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xunming Ji
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Center of Stroke, Beijing Institute for Brain Disorder, Capital Medical University, Beijing 100053, China
| | - Lisha Chang
- North China University of Science and Technology Affiliated Hospital, Tangshan 063000, China.
| |
Collapse
|
13
|
Agrawal R, Sharafkhaneh A, Nambi V, BaHammam A, Razjouyan J. Obstructive sleep apnea modulates clinical outcomes post-acute myocardial infarction: A large longitudinal veterans' dataset report. Respir Med 2023; 211:107214. [PMID: 36924849 PMCID: PMC10122709 DOI: 10.1016/j.rmed.2023.107214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND While the longer-term Obstructive Sleep apnea (OSA)-related intermittent hypoxia (IH) leads to various comorbidities, it has become increasingly evident that OSA confers protective advantages during and after acute myocardial infarction (AMI). We hypothesized in patients who were admitted with acute MI, the presence of OSA is associated with lower in-hospital mortality compared to those without a prior diagnosis of OSA. METHODS In this nationwide retrospective study utilizing Veterans Health Administration records, we included patients hospitalized for MI with a history of sleep disorders from 1999 to 2020. We divided patients into two cohorts: those with OSA and those without OSA. The primary outcome was in-hospital mortality during AMI hospitalization. We analyzed the data using logistic regression and calculated the odds ratio of in-hospital mortality. RESULTS Out of more than four million veterans with any sleep diagnosis, 76,359 patients were hospitalized with a diagnosis of AMI. We observed 30,116 with OSA (age, 64 ± 10 years; BMI, 33 ± 7 kg/m2) and 43,480 without OSA (age, 68 ± 12 years; BMI, 29 ± 6 kg/m2). The aOR of in-patient mortality (n = 333 (1.1%)) was lower in those with OSA (aOR, 0.43; 95% CI, 0.38 to 0.49) compared to without-OSA (n = 1,102, 2.5%). However, the OSA cohort had a higher proportion of the prolonged length of stay (28.1%). CONCLUSIONS Presence of OSA is associated with lower in-hospital mortality among patients admitted for AMI, after adjusting for various demographic and co-morbidity factors. This study highlights the complex relationship between OSA and cardiovascular health and highlights the need for further research in this area.
Collapse
Affiliation(s)
- Ritwick Agrawal
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Pulmonary, Critical Care and Sleep Medicine Section, Medical Care Line, Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Amir Sharafkhaneh
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Pulmonary, Critical Care and Sleep Medicine Section, Medical Care Line, Michael E. DeBakey VA Medical Center, Houston, TX, USA.
| | - Vijay Nambi
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Cardiology Section, Medical Care Line, Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Ahmed BaHammam
- Department of Medicine, University Sleep Disorders Center and Pulmonary Service, King Saud University, Riyadh, Saudi Arabia; Strategic Technologies Program of the National Plan for Sciences and Technology and Innovation in the Kingdom of Saudi Arabia, Saudi Arabia
| | - Javad Razjouyan
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Center for Innovations in Quality, Effectiveness, and Safety, Michael E. DeBakey VA Medical Center, Houston, TX, USA; South Central Mental Illness Research, Education, and Clinical Center, Houston, TX, USA; Big Data Scientist Training Enhancement Program (BD-STEP), VA Office of Research and Development, Washington, DC, USA
| |
Collapse
|
14
|
Gu C, Kong F, Zeng J, Geng X, Sun Y, Chen X. Remote ischemic preconditioning protects against spinal cord ischemia-reperfusion injury in mice by activating NMDAR/AMPK/PGC-1α/SIRT3 signaling. Cell Biosci 2023; 13:57. [PMID: 36927808 PMCID: PMC10018930 DOI: 10.1186/s13578-023-00999-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND To study the protective effects of delayed remote ischemic preconditioning (RIPC) against spinal cord ischemia-reperfusion injury (SCIRI) in mice and determine whether SIRT3 is involved in this protection and portrayed its upstream regulatory mechanisms. METHODS In vivo, WT or SIRT3 global knockout (KO) mice were exposed to right upper and lower limbs RIPC or sham ischemia. After 24 h, the abdominal aorta was clamped for 20 min, then re-perfused for 3 days. The motor function of mice, number of Nissl bodies, apoptotic rate of neurons, and related indexes of oxidative stress in the spinal cord were measured to evaluate for neuroprotective effects. The expression and correlation of SIRT3 and NMDAR were detected by WB and immunofluorescence. In vitro, primary neurons were exacted and OGD/R was performed to simulate SCIRI in vivo. Neuronal damage was assessed by observing neuron morphology, detecting LDH release ratio, and flow cytometry to analyze the apoptosis. MnSOD and CAT enzyme activities, GSH and ROS level were also measured to assess neuronal antioxidant capacity. NMDAR-AMPK-PGC-1α signaling was detected by WB to portray upstream regulatory mechanisms of RIPC regulating SIRT3. RESULTS Compared to the SCIRI mice without RIPC, mice with RIPC displayed improved motor function recovery, a reduced neuronal loss, and enhanced antioxidant capacity. To the contrary, the KO mice did not exhibit any effect of RIPC-induced neuroprotection. Similar results were observed in vitro. Further analyses with spinal cord tissues or primary neurons detected enhanced MnSOD and CAT activities, as well as increased GSH level but decreased MDA or ROS production in the RIPC + I/R mice or NMDA + OGD/R neurons. However, these changes were completely inhibited by the absence of SIRT3. Additionally, NMDAR-AMPK-PGC-1α signaling was activated to upregulate SIRT3 levels, which is essential for RIPC-mediated neuroprotection. CONCLUSIONS RIPC enhances spinal cord ischemia tolerance in a SIRT3-dependent manner, and its induced elevated SIRT3 levels are mediated by the NMDAR-AMPK-PGC-1α signaling pathway. Combined therapy targeting SIRT3 is a promising direction for treating SCIRI.
Collapse
Affiliation(s)
- Changjiang Gu
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, People's Republic of China
| | - Fanqi Kong
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, People's Republic of China
| | - Junkai Zeng
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, People's Republic of China
| | - Xiangwu Geng
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, People's Republic of China
| | - Yanqing Sun
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, 200080, Shanghai, PR China.
| | - Xiongsheng Chen
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, People's Republic of China. .,Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, 200080, Shanghai, PR China.
| |
Collapse
|
15
|
Sirolli V, Piscitani L, Bonomini M. Biomarker-Development Proteomics in Kidney Transplantation: An Updated Review. Int J Mol Sci 2023; 24:ijms24065287. [PMID: 36982359 PMCID: PMC10049725 DOI: 10.3390/ijms24065287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/27/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023] Open
Abstract
Kidney transplantation (KT) is the optimal therapeutic strategy for patients with end-stage renal disease. The key to post-transplantation management is careful surveillance of allograft function. Kidney injury may occur from several different causes that require different patient management approaches. However, routine clinical monitoring has several limitations and detects alterations only at a later stage of graft damage. Accurate new noninvasive biomarker molecules are clearly needed for continuous monitoring after KT in the hope that early diagnosis of allograft dysfunction will lead to an improvement in the clinical outcome. The advent of “omics sciences”, and in particular of proteomic technologies, has revolutionized medical research. Proteomic technologies allow us to achieve the identification, quantification, and functional characterization of proteins/peptides in biological samples such as urine or blood through supervised or targeted analysis. Many studies have investigated proteomic techniques as potential molecular markers discriminating among or predicting allograft outcomes. Proteomic studies in KT have explored the whole transplant process: donor, organ procurement, preservation, and posttransplant surgery. The current article reviews the most recent findings on proteomic studies in the setting of renal transplantation in order to better understand the effective potential of this new diagnostic approach.
Collapse
Affiliation(s)
- Vittorio Sirolli
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS. Annunziata Hospital, 66013 Chieti, Italy
| | - Luca Piscitani
- Nephrology and Dialysis Unit, Department of Medicine, San Salvatore Hospital, 67100 L’Aquila, Italy
| | - Mario Bonomini
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS. Annunziata Hospital, 66013 Chieti, Italy
- Correspondence:
| |
Collapse
|
16
|
Mohamadian M, Parsamanesh N, Chiti H, Sathyapalan T, Sahebkar A. Protective effects of curcumin on ischemia/reperfusion injury. Phytother Res 2022; 36:4299-4324. [PMID: 36123613 DOI: 10.1002/ptr.7620] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/06/2022] [Accepted: 08/24/2022] [Indexed: 12/13/2022]
Abstract
Ischemia/reperfusion (I/R) injury is a term used to describe phenomena connected to the dysfunction of various tissue damage due to reperfusion after ischemic injury. While I/R may result in systemic inflammatory response syndrome or multiple organ dysfunction syndrome, there is still a long way to improve therapeutic outcomes. A number of cellular metabolic and ultrastructural alterations occur by prolonged ischemia. Ischemia increases the expression of proinflammatory gene products and bioactive substances within the endothelium, such as cytokines, leukocytes, and adhesion molecules, even as suppressing the expression of other "protective" gene products and substances, such as thrombomodulin and constitutive nitric oxide synthase (e.g., prostacyclin, nitric oxide [NO]). Curcumin is the primary phenolic pigment derived from turmeric, the powdered rhizome of Curcuma longa. Numerous studies have shown that curcumin has strong antiinflammatory and antioxidant characteristics. It also prevents lipid peroxidation and scavenges free radicals like superoxide anion, singlet oxygen, NO, and hydroxyl. In our study, we highlight the mechanisms of protective effects of curcumin against I/R injury in various organs.
Collapse
Affiliation(s)
- Malihe Mohamadian
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Negin Parsamanesh
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hossein Chiti
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Medicine, The University of Western Australia, Perth, Australia.,Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
17
|
Therapeutic Stimulation of Glycolytic ATP Production for Treating ROS-Mediated Cellular Senescence. Metabolites 2022; 12:metabo12121160. [PMID: 36557198 PMCID: PMC9781421 DOI: 10.3390/metabo12121160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Cellular senescence is conditioned through two interrelated processes, i.e., a reduction in adenosine triphosphate (ATP) and the enhancement of reactive oxygen species (ROS) production levels in mitochondria. ATP shortages primarily influence the energy-intensive synthesis of large biomolecules, such as deoxyribonucleic acid (DNA). In addition, as compared to small biomolecules, large biomolecules are more prone to ROS-mediated damaging effects. Based on the available evidence, we suggest that the stimulation of anaerobic glycolytic ROS-independent ATP production could restrain cellular senescence. Consistent with this notion, non-drug related intermittent hypoxia (IH)-based therapy could be effectively applied in sports medicine, as well as for supporting the physical activity of elderly patients and prophylactics of various age-related disorders. Moreover, drug therapy aiming to achieve the partial blockade of respiratory chain and downstream compensatory glycolysis enhancement could prove to be useful for treating cardiovascular, neurological and hormonal diseases. We maintain that non-drug/drug-related therapeutic interventions applied in combination over the entire lifespan could significantly rejuvenate and prolong a high quality of life for individuals.
Collapse
|
18
|
Belmonte KCD, Holmgren EB, Wills TA, Gidday JM. Epigenetic conditioning induces intergenerational resilience to dementia in a mouse model of vascular cognitive impairment. Alzheimers Dement 2022; 18:1711-1720. [PMID: 35170835 PMCID: PMC9790554 DOI: 10.1002/alz.12616] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/10/2021] [Accepted: 01/10/2022] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Epigenetic stimuli induce beneficial or detrimental changes in gene expression, and consequently, phenotype. Some of these phenotypes can manifest across the lifespan-and even in subsequent generations. Here, we used a mouse model of vascular cognitive impairment and dementia (VCID) to determine whether epigenetically induced resilience to specific dementia-related phenotypes is heritable by first-generation progeny. METHODS Our systemic epigenetic therapy consisted of 2 months of repetitive hypoxic "conditioning" (RHC) prior to chronic cerebral hypoperfusion in adult C57BL/6J mice. Resultant changes in object recognition memory and hippocampal long-term potentiation (LTP) were assessed 3 and 4 months later, respectively. RESULTS Hypoperfusion-induced memory/plasticity deficits were abrogated by RHC. Moreover, similarly robust dementia resilience was documented in untreated cerebral hypoperfused animals derived from RHC-treated parents. CONCLUSIONS Our results in experimental VCID underscore the efficacy of epigenetics-based treatments to prevent memory loss, and demonstrate for the first time the heritability of an induced resilience to dementia.
Collapse
Affiliation(s)
- Krystal Courtney D. Belmonte
- Department of OphthalmologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Department of PhysiologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| | - Eleanor B. Holmgren
- Department of Cell Biology and AnatomyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| | - Tiffany A. Wills
- Department of Cell Biology and AnatomyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Neuroscience Center of ExcellenceLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| | - Jeff M. Gidday
- Department of OphthalmologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Department of PhysiologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Neuroscience Center of ExcellenceLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Department of Biochemistry and Molecular BiologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| |
Collapse
|
19
|
Chen JJ, Lee TH, Kuo G, Huang YT, Chen PR, Chen SW, Yang HY, Hsu HH, Hsiao CC, Yang CH, Lee CC, Chen YC, Chang CH. Strategies for post-cardiac surgery acute kidney injury prevention: A network meta-analysis of randomized controlled trials. Front Cardiovasc Med 2022; 9:960581. [PMID: 36247436 PMCID: PMC9555275 DOI: 10.3389/fcvm.2022.960581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/12/2022] [Indexed: 12/05/2022] Open
Abstract
Objects Cardiac surgery is associated with acute kidney injury (AKI). However, the effects of various pharmacological and non-pharmacological strategies for AKI prevention have not been thoroughly investigated, and their effectiveness in preventing AKI-related adverse outcomes has not been systematically evaluated. Methods Studies from PubMed, Embase, and Medline and registered trials from published through December 2021 that evaluated strategies for preventing post-cardiac surgery AKI were identified. The effectiveness of these strategies was assessed through a network meta-analysis (NMA). The secondary outcomes were prevention of dialysis-requiring AKI, mortality, intensive care unit (ICU) length of stay (LOS), and hospital LOS. The interventions were ranked using the P-score method. Confidence in the results of the NMA was assessed using the Confidence in NMA (CINeMA) framework. Results A total of 161 trials (involving 46,619 participants) and 53 strategies were identified. Eight pharmacological strategies {natriuretic peptides [odds ratio (OR): 0.30, 95% confidence interval (CI): 0.19-0.47], nitroprusside [OR: 0.29, 95% CI: 0.12-0.68], fenoldopam [OR: 0.36, 95% CI: 0.17-0.76], tolvaptan [OR: 0.35, 95% CI: 0.14-0.90], N-acetyl cysteine with carvedilol [OR: 0.37, 95% CI: 0.16-0.85], dexmedetomidine [OR: 0.49, 95% CI: 0.32-0.76;], levosimendan [OR: 0.56, 95% CI: 0.37-0.84], and erythropoietin [OR: 0.62, 95% CI: 0.41-0.94]} and one non-pharmacological intervention (remote ischemic preconditioning, OR: 0.76, 95% CI: 0.63-0.92) were associated with a lower incidence of post-cardiac surgery AKI with moderate to low confidence. Among these nine strategies, five (fenoldopam, erythropoietin, natriuretic peptides, levosimendan, and remote ischemic preconditioning) were associated with a shorter ICU LOS, and two (natriuretic peptides [OR: 0.30, 95% CI: 0.15-0.60] and levosimendan [OR: 0.68, 95% CI: 0.49-0.95]) were associated with a lower incidence of dialysis-requiring AKI. Natriuretic peptides were also associated with a lower risk of mortality (OR: 0.50, 95% CI: 0.29-0.86). The results of a sensitivity analysis support the robustness and effectiveness of natriuretic peptides and dexmedetomidine. Conclusion Nine potentially effective strategies were identified. Natriuretic peptide therapy was the most effective pharmacological strategy, and remote ischemic preconditioning was the only effective non-pharmacological strategy. Preventive strategies might also help prevent AKI-related adverse outcomes. Additional studies are required to explore the optimal dosages and protocols for potentially effective AKI prevention strategies.
Collapse
Affiliation(s)
- Jia-Jin Chen
- Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | | | - George Kuo
- Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Nephrology, Kidney Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yen-Ta Huang
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Rung Chen
- Department of Anesthesiology, Mackay Memorial Hospital, Taipei, Taiwan
| | - Shao-Wei Chen
- Department of Cardiothoracic and Vascular Surgery, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Huang-Yu Yang
- Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Nephrology, Kidney Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Hsiang-Hao Hsu
- Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Nephrology, Kidney Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ching-Chung Hsiao
- Department of Nephrology, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan
| | - Chia-Hung Yang
- Department of Cardiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Cheng-Chia Lee
- Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Nephrology, Kidney Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yung-Chang Chen
- Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Nephrology, Kidney Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chih-Hsiang Chang
- Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Nephrology, Kidney Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
20
|
Wang Y, Leak RK, Cao G. Microglia-mediated neuroinflammation and neuroplasticity after stroke. Front Cell Neurosci 2022; 16:980722. [PMID: 36052339 PMCID: PMC9426757 DOI: 10.3389/fncel.2022.980722] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 07/22/2022] [Indexed: 11/13/2022] Open
Abstract
Stroke remains a major cause of long-term disability and mortality worldwide. The immune system plays an important role in determining the condition of the brain following stroke. As the resident innate immune cells of the central nervous system, microglia are the primary responders in a defense network covering the entire brain parenchyma, and exert various functions depending on dynamic communications with neurons, astrocytes, and other neighboring cells under both physiological or pathological conditions. Microglia activation and polarization is crucial for brain damage and repair following ischemic stroke, and is considered a double-edged sword for neurological recovery. Microglia can exist in pro-inflammatory states and promote secondary brain damage, but they can also secrete anti-inflammatory cytokines and neurotrophic factors and facilitate recovery following stroke. In this review, we focus on the role and mechanisms of microglia-mediated neuroinflammation and neuroplasticity after ischemia and relevant potential microglia-based interventions for stroke therapy.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
- *Correspondence: Guodong Cao Yuan Wang
| | - Rehana K. Leak
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Guodong Cao
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, United States
- *Correspondence: Guodong Cao Yuan Wang
| |
Collapse
|
21
|
González-Candia A, Candia AA, Paz A, Mobarec F, Urbina-Varela R, del Campo A, Herrera EA, Castillo RL. Cardioprotective Antioxidant and Anti-Inflammatory Mechanisms Induced by Intermittent Hypobaric Hypoxia. Antioxidants (Basel) 2022; 11:antiox11061043. [PMID: 35739940 PMCID: PMC9220055 DOI: 10.3390/antiox11061043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/25/2022] Open
Abstract
More than 80 million people live and work (in a chronic or intermittent form) above 2500 masl, and 35 million live in the Andean Mountains. Furthermore, in Chile, it is estimated that 100,000 people work in high-altitude shifts, where stays in the lowlands are interspersed with working visits in the highlands. Acute exposure to high altitude has been shown to induce oxidative stress in healthy human lowlanders due to increased free radical formation and decreased antioxidant capacity. However, intermittent hypoxia (IH) induces preconditioning in animal models, generating cardioprotection. Here, we aim to describe the responses of a cardiac function to four cycles of intermittent hypobaric hypoxia (IHH) in a rat model. The twelve adult Wistar rats were randomly divided into two equal groups, a four-cycle of IHH and a normobaric hypoxic control. Intermittent hypoxia was induced in a hypobaric chamber in four continuous cycles (1 cycle = 4 days of hypoxia + 4 days of normoxia), reaching a barometric pressure equivalent to 4600 m of altitude (428 Torr). At the end of the fourth cycle, cardiac structural and functional variables were also determined by echocardiography; furthermore, cardiac oxidative stress biomarkers (4-Hydroxynonenal, HNE; nitrotyrosine, NT), antioxidant enzymes, and NLRP3 inflammasome panel expression are also determined. Our results show a higher ejection and a shortening fraction of the left ventricle function by the end of the fourth cycle. Furthermore, cardiac tissue presented a decreased expression of antioxidant proteins. However, a decrease in IL-1β, TNF-αn, and oxidative stress markers is observed in IHH compared to normobaric hypoxic controls. Non-significant differences were found in protein levels of NLRP3 and caspase-1. IHH exposure determines structural and functional heart changes. These findings suggest that initial states of IHH are beneficial for cardiovascular function and protection.
Collapse
Affiliation(s)
| | - Alejandro A. Candia
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago 7500922, Chile; (A.A.C.); (A.P.); (F.M.)
- Department for the Woman and Newborn Health Promotion, Universidad de Chile, Santiago 7500922, Chile
| | - Adolfo Paz
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago 7500922, Chile; (A.A.C.); (A.P.); (F.M.)
| | - Fuad Mobarec
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago 7500922, Chile; (A.A.C.); (A.P.); (F.M.)
| | - Rodrigo Urbina-Varela
- Laboratorio de Fisiología y Bioenergética Celular, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile; (R.U.-V.); (A.d.C.)
| | - Andrea del Campo
- Laboratorio de Fisiología y Bioenergética Celular, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile; (R.U.-V.); (A.d.C.)
| | - Emilio A. Herrera
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago 7500922, Chile; (A.A.C.); (A.P.); (F.M.)
- International Center for Andean Studies (INCAS), University of Chile, Putre 1070000, Chile
- Correspondence: (E.A.H.); or (R.L.C.); Tel.: +56-982-337-566 (R.L.C.)
| | - Rodrigo L. Castillo
- Departamento de Medicina Interna Oriente, Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile
- Unidad de Paciente Crítico, Hospital del Salvador, Santiago 7500922, Chile
- Correspondence: (E.A.H.); or (R.L.C.); Tel.: +56-982-337-566 (R.L.C.)
| |
Collapse
|
22
|
Roles of Nitric Oxide in Brain Ischemia and Reperfusion. Int J Mol Sci 2022; 23:ijms23084243. [PMID: 35457061 PMCID: PMC9028809 DOI: 10.3390/ijms23084243] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/08/2022] [Accepted: 04/08/2022] [Indexed: 01/27/2023] Open
Abstract
Brain ischemia and reperfusion (I/R) is one of the most severe clinical manifestations of ischemic stroke, placing a significant burden on both individuals and society. The only FDA-approved clinical treatment for ischemic stroke is tissue plasminogen activator (t-PA), which rapidly restores cerebral blood flow but can have severe side effects. The complex pathological process of brain I/R has been well-established in the past few years, including energy metabolism disorders, cellular acidosis, doubling of the synthesis or release of excitotoxic amino acids, intracellular calcium homeostasis, free radical production, and activation of apoptotic genes. Recently, accumulating evidence has shown that NO may be strongly related to brain I/R and involved in complex pathological processes. This review focuses on the role of endogenous NO in pathological processes in brain I/R, including neuronal cell death and blood brain barrier disruption, to explore how NO impacts specific signaling cascades and contributes to brain I/R injury. Moreover, NO can rapidly react with superoxide to produce peroxynitrite, which may also mediate brain I/R injury, which is discussed here. Finally, we reveal several therapeutic approaches strongly associated with NO and discuss their potential as a clinical treatment for ischemic stroke.
Collapse
|
23
|
Serebrovska ZO, Xi L, Tumanovska LV, Shysh AM, Goncharov SV, Khetsuriani M, Kozak TO, Pashevin DA, Dosenko VE, Virko SV, Kholin VA, Grib ON, Utko NA, Egorov E, Polischuk AO, Serebrovska TV. Response of Circulating Inflammatory Markers to Intermittent Hypoxia-Hyperoxia Training in Healthy Elderly People and Patients with Mild Cognitive Impairment. Life (Basel) 2022; 12:life12030432. [PMID: 35330183 PMCID: PMC8953753 DOI: 10.3390/life12030432] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 12/28/2022] Open
Abstract
Intermittent hypoxia-hyperoxia training (IHHT) is a non-pharmacological therapeutic modality for management of some chronic- and age-related pathologies, such as Alzheimer’s disease (AD). Our previous studies demonstrated significant improvement of cognitive function after IHHT in the patients with mild cognitive impairment (MCI). The present study further investigated the effects of IHHT on pro-inflammatory factors in healthy elderly individuals and patients with early signs of AD. Twenty-nine subjects (13 healthy subjects without signs of cognitive impairment syndrome and 16 patients diagnosed with MCI; age 52 to 76 years) were divided into four groups: Healthy+Sham (n = 7), Healthy+IHHT (n = 6), MCI+Sham (n = 6), and MCI+IHHT (n = 10). IHHT was carried out 5 days per week for 3 weeks (total 15 sessions), and each daily session included 4 cycles of 5-min hypoxia (12% FIO2) and 3-min hyperoxia (33% FIO2). Decline in cognitive function indices was observed initially in both MCI+Sham and MCI+IHHT groups. The sham training did not alter any of the parameters, whereas IHHT resulted in improvement in latency of cognitive evoked potentials, along with elevation in APP110, GDF15 expression, and MMP9 activity in both healthy subjects and those with MCI. Increased MMP2 activity, HMGB1, and P-selectin expression and decreased NETs formation and Aβ expression were also observed in the MCI+IHHT group. There was a negative correlation between MoCA score and the plasma GDF15 expression (R = −0.5799, p < 0.05) before the initiation of IHHT. The enhanced expression of GDF15 was also associated with longer latency of the event-related potentials P330 and N200 (R = 0.6263, p < 0.05 and R = 0.5715, p < 0.05, respectively). In conclusion, IHHT upregulated circulating levels of some inflammatory markers, which may represent potential triggers for cellular adaptive reprogramming, leading to therapeutic effects against cognitive dysfunction and neuropathological changes during progression of AD. Further investigation is needed to clarify if there is a causative relationship between the improved cognitive function and the elevated inflammatory markers following IHHT.
Collapse
Affiliation(s)
- Zoya O. Serebrovska
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine; (L.V.T.); (A.M.S.); (S.V.G.); (M.K.); (T.O.K.); (D.A.P.); (V.E.D.); (A.O.P.); (T.V.S.)
- Correspondence: (Z.O.S.); (L.X.)
| | - Lei Xi
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298-0204, USA
- Correspondence: (Z.O.S.); (L.X.)
| | - Lesya V. Tumanovska
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine; (L.V.T.); (A.M.S.); (S.V.G.); (M.K.); (T.O.K.); (D.A.P.); (V.E.D.); (A.O.P.); (T.V.S.)
| | - Angela M. Shysh
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine; (L.V.T.); (A.M.S.); (S.V.G.); (M.K.); (T.O.K.); (D.A.P.); (V.E.D.); (A.O.P.); (T.V.S.)
| | - Sergii V. Goncharov
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine; (L.V.T.); (A.M.S.); (S.V.G.); (M.K.); (T.O.K.); (D.A.P.); (V.E.D.); (A.O.P.); (T.V.S.)
| | - Michael Khetsuriani
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine; (L.V.T.); (A.M.S.); (S.V.G.); (M.K.); (T.O.K.); (D.A.P.); (V.E.D.); (A.O.P.); (T.V.S.)
| | - Taisia O. Kozak
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine; (L.V.T.); (A.M.S.); (S.V.G.); (M.K.); (T.O.K.); (D.A.P.); (V.E.D.); (A.O.P.); (T.V.S.)
| | - Denis A. Pashevin
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine; (L.V.T.); (A.M.S.); (S.V.G.); (M.K.); (T.O.K.); (D.A.P.); (V.E.D.); (A.O.P.); (T.V.S.)
| | - Victor E. Dosenko
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine; (L.V.T.); (A.M.S.); (S.V.G.); (M.K.); (T.O.K.); (D.A.P.); (V.E.D.); (A.O.P.); (T.V.S.)
| | - Sergii V. Virko
- Lashkariov Institute of Semiconductor Physics, National Academy of Sciences, 41 Nauki Ave., 03028 Kyiv, Ukraine;
| | - Viktor A. Kholin
- Department of Age Physiology and Pathology of Nervous System, Chebotarev Institute of Gerontology NAMS of Ukraine, 04114 Kyiv, Ukraine; (V.A.K.); (O.N.G.); (N.A.U.)
| | - Oksana N. Grib
- Department of Age Physiology and Pathology of Nervous System, Chebotarev Institute of Gerontology NAMS of Ukraine, 04114 Kyiv, Ukraine; (V.A.K.); (O.N.G.); (N.A.U.)
| | - Natalie A. Utko
- Department of Age Physiology and Pathology of Nervous System, Chebotarev Institute of Gerontology NAMS of Ukraine, 04114 Kyiv, Ukraine; (V.A.K.); (O.N.G.); (N.A.U.)
| | - Egor Egorov
- CELLGYM Technologies GmbH, 14193 Berlin, Germany;
| | - Anna O. Polischuk
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine; (L.V.T.); (A.M.S.); (S.V.G.); (M.K.); (T.O.K.); (D.A.P.); (V.E.D.); (A.O.P.); (T.V.S.)
| | - Tetiana V. Serebrovska
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, 01024 Kyiv, Ukraine; (L.V.T.); (A.M.S.); (S.V.G.); (M.K.); (T.O.K.); (D.A.P.); (V.E.D.); (A.O.P.); (T.V.S.)
| |
Collapse
|
24
|
Yoshihara T, Matsumura N, Tamura T, Shiozaki S, Tobita S. Intracellular and Intravascular Oxygen Sensing of Pancreatic Tissues Based on Phosphorescence Lifetime Imaging Microscopy Using Lipophilic and Hydrophilic Iridium(III) Complexes. ACS Sens 2022; 7:545-554. [PMID: 35113520 DOI: 10.1021/acssensors.1c02379] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Simultaneous imaging of intracellular and blood oxygen levels in tissues provides valuable information on the dynamic behavior of molecular oxygen (O2) in normal and diseased tissues. Here, to achieve this goal, we developed green-emitting intracellular O2 probes based on the Ir(III) complex, PPY (tris(2-phenylpyridinato)iridium(III)), and investigated the possibility of multicolor O2 imaging by co-staining tissues with a red-emitting intravascular probe BTP-PEG48. The newly synthesized complexes possess modified 2-phenylpyridinato ligand(s) with a cationic or hydrophilic substituent, such as a dimethylamino group, triphenylphosphonium cation, or hydroxy group, in order to enhance cellular uptake efficiency. The photophysical and cellular properties of these complexes were systematically investigated to evaluate their ability as O2 probes. Among these complexes, PPYDM and PPY2OH, which have a dimethylamino group and two hydroxy groups, respectively, exhibited much higher cellular uptake efficiencies compared with PPY and showed high O2 sensitivity in HeLa cells. Phosphorescence lifetime imaging microscopy (PLIM) measurements of HeLa cells co-stained with PPYDM and hydrophilic BTP-PEG48 allowed for the evaluation of intracellular and extracellular O2 levels in cell culture. We took PLIM images of the pancreas following intravenous administration of PPYDM and BTP-PEG48 into anesthetized mice. The PLIM measurements using these probes allowed simultaneous O2 imaging of acinar cells and capillaries in the pancreas with cellular-level resolution. From the phosphorescence lifetimes of PPYDM and BTP-PEG48 and the calibration curves evaluated in rat pancreatic acinar cells and blood plasma, we found that the average oxygen partial pressures of acinar cells and capillaries were almost equal at about 30 mmHg.
Collapse
Affiliation(s)
- Toshitada Yoshihara
- Department of Chemistry and Chemical Biology, Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-cho, Kiryu, Gunma 376-8515, Japan
| | - Nao Matsumura
- Department of Chemistry and Chemical Biology, Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-cho, Kiryu, Gunma 376-8515, Japan
| | - Takuto Tamura
- Department of Chemistry and Chemical Biology, Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-cho, Kiryu, Gunma 376-8515, Japan
| | - Shuichi Shiozaki
- Department of Chemistry and Chemical Biology, Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-cho, Kiryu, Gunma 376-8515, Japan
| | - Seiji Tobita
- Department of Chemistry and Chemical Biology, Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-cho, Kiryu, Gunma 376-8515, Japan
| |
Collapse
|
25
|
Ghozy S, Kacimi SEO, Elfil M, Sobeeh MG, Reda A, Kallmes KM, Rabinstein AA, Holmes DR, Brinjikji W, Kadirvel R, Kallmes DF. Transient Ischemic Attacks Preceding Ischemic Stroke and the Possible Preconditioning of the Human Brain: A Systematic Review and Meta-Analysis. Front Neurol 2021; 12:755167. [PMID: 34899573 PMCID: PMC8652229 DOI: 10.3389/fneur.2021.755167] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/25/2021] [Indexed: 01/10/2023] Open
Abstract
Stroke is a leading cause of mortality and disability worldwide. Transient ischemic attack (TIA) is defined as transient brain ischemia with temporary neurological deficits. In animal models, prior TIA seems to enhance brain ischemic tolerance to withstand further ischemic events, which might be explained by brain preconditioning. Thus, this review aims to formulate evidence of whether TIAs can induce positive preconditioning and enhance the functional outcomes in patients suffering from subsequent ischemic strokes. Five databases were searched (PubMed, Embase, SAGE, Web of Science, and Scopus), and twelve studies were included in the quantitative analysis. Studies were eligible when comparing patients with acute ischemic stroke (AIS) and previous TIA with those with AIS without TIA. Comparisons included the National Institute of Health Stroke Scale (NIHSS) score at admission and 7 days from the stroke event, modified Rankin score (mRS), and Trial of ORG 10,172 in Acute Stroke Treatment (TOAST) classification. Odds ratio (OR), mean difference (MD), and 95% confidence interval (CI) were used to describe our results using the random effect model. Our results revealed that patients with stroke and prior TIAs had lower NIHSS scores at admission than those without prior TIAs. However, the NIHSS score was not significantly different between the two groups at 7 days. Furthermore, there was no statistically significant difference between both groups in terms of mortality. Despite the differences in the admission mRS score groups, patients with prior TIAs had lower mRS scores at discharge.
Collapse
Affiliation(s)
- Sherief Ghozy
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | | | - Mohammed Elfil
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Mohamed Gomaa Sobeeh
- Faculty of Physical Therapy, Cairo University, Cairo, Egypt.,Faculty of Physical Therapy, Sinai University, Cairo, Egypt
| | - Abdullah Reda
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Kevin M Kallmes
- Nested Knowledge, St. Paul, MN, United States.,Superior Medical Experts, St. Paul, MN, United States
| | - Alejandro A Rabinstein
- Department of Neurology and Neurocritical Care, Mayo Clinic, Rochester, MN, United States
| | - David R Holmes
- Department of Cardiovascular Diseases and Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Waleed Brinjikji
- Department of Radiology, Mayo Clinic, Rochester, MN, United States.,Department of Neurosurgery, Mayo Clinic Rochester, Rochester, MN, United States
| | | | - David F Kallmes
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
26
|
Lu N, Bai R, Liu B, Cheng W, Wu Z. Attenuation of the cytoprotection induced by hypoxic preconditioning upon transfection with BNIP3-siRNA in human neuroblastoma SH-SY5Y cells. Int J Neurosci 2021:1-10. [PMID: 34871150 DOI: 10.1080/00207454.2021.2015349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
PURPOSE The aim of this study was to investigate the functional role of hypoxic preconditioning (HPC) in human neuroblastoma cells. METHODS BNIP3 small-interfering RNA (BNIP3-siRNA) sequence was synthesized and used to transfect human neuroblastoma SH-SY5Y cell lines. Thereafter, BNIP3 expression at mRNA and protein levels and its effects on the cell proliferation were analyzed. The most effective pair of siRNA was selected to knockdown the expression level of BNIP3. Moreover, the effects of HPC on oxygen-glucose deprivation/reperfusion (OGD/R)-induced apoptosis and autophagy in SH-SY5Y cells were explored to further reveal the possible mechanisms underlying HPC. RESULTS BNIP3-siRNA attenuated the protective effects of HPC by decreasing the cell viability, increasing the enzymatic activity of caspase-3 and 9, increasing the rate of apoptosis, and increasing the protein expression level of activated caspase-3. Additionally, BNIP3-siRNA had no significant influence on the expression level of HIF-1α induced by HPC, while it substantially inhibited HPC-induced BNIP3/Beclin1 and autophagy. CONCLUSIONS HPC promoted autophagy through regulating BNIP3 to reduce OGD/R.
Collapse
Affiliation(s)
- Na Lu
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Ruiying Bai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Bo Liu
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Weijia Cheng
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Zekun Wu
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
27
|
Abstract
Cerebral ischemic injury may lead to a series of serious brain diseases, death or different degrees of disability. Hypoxia-inducible factor-1α (HIF-1α) is an oxygen-sensitive transcription factor, which mediates the adaptive metabolic response to hypoxia and serves a key role in cerebral ischemia. HIF-1α is the main molecule that responds to hypoxia. HIF-1α serves an important role in the development of cerebral ischemia by participating in numerous processes, including metabolism, proliferation and angiogenesis. The present review focuses on the endogenous protective mechanism of cerebral ischemia and elaborates on the role of HIF-1α in cerebral ischemia. In addition, it focuses on cerebral ischemia interventions that act on the HIF-1α target, including biological factors, non-coding RNA, hypoxic-ischemic preconditioning and drugs, and expands upon the measures to strengthen the endogenous compensatory response to support HIF-1α as a therapeutic target, thus providing novel suggestions for the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Peiliang Dong
- Institute of Traditional Chinese Medicine, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Qingna Li
- College of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Hua Han
- College of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| |
Collapse
|
28
|
Effect of Intrahippocampal Administration of α7 Subtype Nicotinic Receptor Agonist PNU-282987 and Its Solvent Dimethyl Sulfoxide on the Efficiency of Hypoxic Preconditioning in Rats. Molecules 2021; 26:molecules26237387. [PMID: 34885970 PMCID: PMC8659180 DOI: 10.3390/molecules26237387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/27/2021] [Accepted: 12/01/2021] [Indexed: 11/17/2022] Open
Abstract
We have previously suggested a key role of the hippocampus in the preconditioning action of moderate hypobaric hypoxia (HBH). The preconditioning efficiency of HBH is associated with acoustic startle prepulse inhibition (PPI). In rats with PPI > 40%, HBH activates the cholinergic projections of hippocampus, and PNU-282987, a selective agonist of α7 nicotinic receptors (α7nAChRs), reduces the HBH efficiency and potentiating effect on HBH of its solvent dimethyl sulfoxide (DMSO, anticholinesterase agent) when administered intraperitoneally. In order to validate the hippocampus as a key structure in the mechanism of hypoxic preconditioning and research a significance of α7nAChR activation in the hypoxic preconditioning, we performed an in vivo pharmacological study of intrahippocampal injections of PNU-282987 into the CA1 area on HBH efficiency in rats with PPI ≥ 40%. We found that PNU-282987 (30 μM) reduced HBH efficiency as with intraperitoneal administration, while DMSO (0.05%) still potentiated this effect. Thus, direct evidence of the key role of the hippocampus in the preconditioning effect of HBH and some details of this mechanism were obtained in rats with PPI ≥ 40%. The activation of α7nAChRs is not involved in the cholinergic signaling initiated by HBH or DMSO via any route of administration. Possible ways of the potentiating action of DMSO on HBH efficiency and its dependence on α7nAChRs are discussed.
Collapse
|
29
|
Gatto M, Mota GAF, Pagan LU, Gomes MJ, Okoshi MP. Pré-Condicionamento na Lesão por Isquemia-Reperfusão. Arq Bras Cardiol 2021; 117:1145-1146. [PMID: 35613171 PMCID: PMC8757153 DOI: 10.36660/abc.20210908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
30
|
N-myc Downstream-Regulated Gene 2 (NDRG2) Function as a Positive Regulator of Apoptosis: A New Insight into NDRG2 as a Tumor Suppressor. Cells 2021; 10:cells10102649. [PMID: 34685629 PMCID: PMC8534062 DOI: 10.3390/cells10102649] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022] Open
Abstract
N-myc downstream-regulated gene 2 (NDRG2) is a tumor suppressor gene that increases tumor sensitivity to anticancer drugs, slows tumor progression, and inhibits metastasis. NDRG2 is suppressed in various aggressive tumor positions, whereas NDRG2 expression is associated with patient prognosis, such as an improved survival rate. In this review, we summarize the tumor suppressor mechanism of NDRG2 and provide information on the function of NDRG2 concerning the susceptibility of cells to apoptosis. NDRG2 increases the susceptibility to apoptosis in various physiological environments of cells, such as development, hypoxia, nutrient deprivation, and cancer drug treatment. Although the molecular and cell biological mechanisms of NDRG2 have not been fully elucidated, we provide information on the mechanisms of NDRG2 in relation to apoptosis in various environments. This review can assist the design of research regarding NDRG2 function and suggests the potential of NDRG2 as a molecular target for cancer patients.
Collapse
|
31
|
Li Y, Huang Y, Cheng X, He Y, Hu X. Whole body hypoxic preconditioning-mediated multiorgan protection in db/db mice via nitric oxide-BDNF-GSK-3β-Nrf2 signaling pathway. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2021; 25:281-296. [PMID: 34187947 PMCID: PMC8255126 DOI: 10.4196/kjpp.2021.25.4.281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 11/15/2022]
Abstract
The beneficial effects of hypoxic preconditioning are abolished in the diabetes. The present study was designed to investigate the protective effects and mechanisms of repeated episodes of whole body hypoxic preconditioning (WBHP) in db/db mice. The protective effects of preconditioning were explored on diabetesinduced vascular dysfunction, cognitive impairment and ischemia-reperfusion (IR)-induced increase in myocardial injury. Sixteen-week old db/db (diabetic) and C57BL/6 (non-diabetic) mice were employed. There was a significant impairment in cognitive function (Morris Water Maze test), endothelial function (acetylcholineinduced relaxation in aortic rings) and a significant increase in IR-induced heart injury (Langendorff apparatus) in db/db mice. WBHP stimulus was given by exposing mice to four alternate cycles of low (8%) and normal air O2 for 10 min each. A single episode of WBHP failed to produce protection; however, two and three episodes of WBHP significantly produced beneficial effects on the heart, brain and blood vessels. There was a significant increase in the levels of brain-derived neurotrophic factor (BDNF) and nitric oxide (NO) in response to 3 episodes of WBHP. Moreover, pretreatment with the BDNF receptor, TrkB antagonist (ANA-12) and NO synthase inhibitor (LNAME) attenuated the protective effects imparted by three episodes of WBHP. These pharmacological agents abolished WBHP-induced restoration of p-GSK-3β/GSK-3β ratio and Nrf2 levels in IR-subjected hearts. It is concluded that repeated episodes of WHBP attenuate cognitive impairment, vascular dysfunction and enhancement in IRinduced myocardial injury in diabetic mice be due to increase in NO and BDNF levels that may eventually activate GSK-3β and Nrf2 signaling pathway to confer protection.
Collapse
Affiliation(s)
- Yuefang Li
- Cadre Ward the No.901 Hospital of the Joint Logistics Support Unit of the Chinese People's Liberation Army, Hefei, Anhui 230031, P.R. China
| | - Yan Huang
- Cadre Ward the No.901 Hospital of the Joint Logistics Support Unit of the Chinese People's Liberation Army, Hefei, Anhui 230031, P.R. China
| | - Xi Cheng
- Cadre Ward the No.901 Hospital of the Joint Logistics Support Unit of the Chinese People's Liberation Army, Hefei, Anhui 230031, P.R. China
| | - Youjun He
- Cadre Ward the No.901 Hospital of the Joint Logistics Support Unit of the Chinese People's Liberation Army, Hefei, Anhui 230031, P.R. China
| | - Xin Hu
- Cadre Ward the No.901 Hospital of the Joint Logistics Support Unit of the Chinese People's Liberation Army, Hefei, Anhui 230031, P.R. China
| |
Collapse
|
32
|
Hansen LF, Nielsen NSK, Christoffersen LC, Kruuse C. Translational challenges of remote ischemic conditioning in ischemic stroke - a systematic review. Ann Clin Transl Neurol 2021; 8:1720-1729. [PMID: 34133841 PMCID: PMC8351389 DOI: 10.1002/acn3.51405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/20/2021] [Accepted: 05/19/2021] [Indexed: 12/27/2022] Open
Abstract
Remote ischemic conditioning (RIC) has well‐established cardioprotective effects in preclinical studies and promising results in preclinical stroke research. Effective translation from preclinical studies to clinical trials has yet to be accomplished, perhaps because of the use of multiple applications of RIC (e.g., pre‐, per‐, or post‐conditioning) in preclinical studies by both invasive and non‐invasive protocols, some of which not clinically applicable. Our systematic review conformed to PRISMA guidelines and addressed differences in clinically relevant RIC applications and outcomes between preclinical and clinical studies. We retrieved a total of 30 studies (8 human; 22 animal) that met the inclusion criteria of testing clinically relevant procedures; namely, non‐invasive and per‐ or post‐conditioning protocols. Per‐conditioning was applied in 6 animal and 3 human studies, post‐conditioning was applied in 16 animal and 5 human studies, and both conditioning methods were applied in 2 animal studies. Application of RIC varied between human and animal studies regarding initiation, duration, repetition, and number of limbs included. Study designs did not systematically apply blinding, randomization, or placebo controls. On only a few occasions did preclinical studies include animals with clinically relevant comorbidities. Clinical trials were challenged by not completing the intended number of RIC cycles or addressing this deficit in the data analysis. Consistency and transferability of methods used for positive animal studies and subsequent human studies are essential for the optimal translation of results. Consensus on preclinical and clinical RIC procedures should be reached for a full understanding of the possible beneficial effects of RIC treatment in stroke.
Collapse
Affiliation(s)
- Line Fuglsang Hansen
- Department of Neurology, Neurovascular Research Unit, Herlev Gentofte Hospital, Copenhagen, Denmark.,Department of Anesthesiology and Intensive Care, Holbaek Hospital, Holbaek, Denmark
| | - Nicholine S K Nielsen
- Department of Neurology, Neurovascular Research Unit, Herlev Gentofte Hospital, Copenhagen, Denmark
| | | | - Christina Kruuse
- Department of Neurology, Neurovascular Research Unit, Herlev Gentofte Hospital, Copenhagen, Denmark.,Department. of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Remote Ischemic Conditioning in Emergency Medicine-Clinical Frontiers and Research Opportunities. Shock 2021; 53:269-276. [PMID: 32045394 DOI: 10.1097/shk.0000000000001362] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Time-critical acute ischemic conditions such as ST-elevation myocardial infarction and acute ischemic stroke are staples in Emergency Medicine practice. While timely reperfusion therapy is a priority, the resultant acute ischemia/reperfusion injury contributes to significant mortality and morbidity. Among therapeutics targeting ischemia/reperfusion injury (IRI), remote ischemic conditioning (RIC) has emerged as the most promising.RIC, which consists of repetitive inflation and deflation of a pneumatic cuff on a limb, was first demonstrated to have protective effect on IRI through various neural and humoral mechanisms. Its attractiveness stems from its simplicity, low-cost, safety, and efficacy, while at the same time it does not impede reperfusion treatment. There is now good evidence for RIC as an effective adjunct to reperfusion in ST-elevation myocardial infarction patients for improving clinical outcomes. For other applications such as acute ischemic stroke, subarachnoid hemorrhage, traumatic brain injury, cardiac arrest, and spinal injury, there is varying level of evidence.This review aims to describe the RIC phenomenon, briefly recount its historical development, and appraise the experimental and clinical evidence for RIC in selected emergency conditions. Finally, it describes the practical issues with RIC clinical application and research in Emergency Medicine.
Collapse
|
34
|
Yang X, Lu W, Hopper CP, Ke B, Wang B. Nature's marvels endowed in gaseous molecules I: Carbon monoxide and its physiological and therapeutic roles. Acta Pharm Sin B 2021; 11:1434-1445. [PMID: 34221861 PMCID: PMC8245769 DOI: 10.1016/j.apsb.2020.10.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 08/03/2020] [Accepted: 09/07/2020] [Indexed: 02/08/2023] Open
Abstract
Nature has endowed gaseous molecules such as O2, CO2, CO, NO, H2S, and N2 with critical and diverse roles in sustaining life, from supplying energy needed to power life and building blocks for life's physical structure to mediating and coordinating cellular functions. In this article, we give a brief introduction of the complex functions of the various gaseous molecules in life and then focus on carbon monoxide as a specific example of an endogenously produced signaling molecule to highlight the importance of this class of molecules. The past twenty years have seen much progress in understanding CO's mechanism(s) of action and pharmacological effects as well as in developing delivery methods for easy administration. One remarkable trait of CO is its pleiotropic effects that have few parallels, except perhaps its sister gaseous signaling molecules such as nitric oxide and hydrogen sulfide. This review will delve into the sophistication of CO-mediated signaling as well as its validated pharmacological functions and possible therapeutic applications.
Collapse
Affiliation(s)
- Xiaoxiao Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Wen Lu
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Christopher P. Hopper
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
- Institut für Experimentelle Biomedizin, Universitätsklinikum Würzburg, Würzburg, Bavaria 97080, Germany
| | - Bowen Ke
- Department of Anesthesiology, West China Hospital, Chengdu 610041, China
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
35
|
Utilizing Developmentally Essential Secreted Peptides Such as Thymosin Beta-4 to Remind the Adult Organs of Their Embryonic State-New Directions in Anti-Aging Regenerative Therapies. Cells 2021; 10:cells10061343. [PMID: 34071596 PMCID: PMC8228050 DOI: 10.3390/cells10061343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/07/2021] [Accepted: 05/20/2021] [Indexed: 01/10/2023] Open
Abstract
Our dream of defeating the processes of aging has occupied the curious and has challenged scientists globally for hundreds of years. The history is long, and sadly, the solution is still elusive. Our endeavors to reverse the magnitude of damaging cellular and molecular alterations resulted in only a few, yet significant advancements. Furthermore, as our lifespan increases, physicians are facing more mind-bending questions in their routine practice than ever before. Although the ultimate goal is to successfully treat the body as a whole, steps towards regenerating individual organs are even considered significant. As our initial approach to enhance the endogenous restorative capacity by delivering exogenous progenitor cells appears limited, we propose, utilizing small molecules critical during embryonic development may prove to be a powerful tool to increase regeneration and to reverse the processes associated with aging. In this review, we introduce Thymosin beta-4, a 43aa secreted peptide fulfilling our hopes and capable of numerous regenerative achievements via systemic administration in the heart. Observing the broad capacity of this small, secreted peptide, we believe it is not the only molecule which nature conceals to our benefit. Hence, the discovery and postnatal administration of developmentally relevant agents along with other approaches may result in reversing the aging process.
Collapse
|
36
|
The hormetic dose-response mechanism: Nrf2 activation. Pharmacol Res 2021; 167:105526. [DOI: 10.1016/j.phrs.2021.105526] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022]
|
37
|
Lu Y, Bian J, Kan H, Ding W, Wang D, Wang X, Luo Q, Wu X, Zhu L. Intermittent hypoxia preconditioning protects WRL68 cells against oxidative injury: Involvement of the PINK1/Parkin-mediated mitophagy regulated by nuclear respiratory factor 1. Mitochondrion 2021; 59:113-122. [PMID: 33933661 DOI: 10.1016/j.mito.2021.04.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 04/04/2021] [Accepted: 04/26/2021] [Indexed: 01/22/2023]
Abstract
The protective effect of intermittent hypoxia (IH) preconditioning against oxidative injury in hepatic cells was investigated and the involvement of the PINK1/Parkin-mediated mitophagy regulated by nuclear respiratory factor 1 (NRF-1) was evaluated. The results showed that IH preconditioning protected HepG2 cells against oxygen and glucose deprivation/reperfusion (OGD/Rep)-induced injury and protected WRL68 cells against H2O2 or AMA-induced oxidative injury. IH preconditioning up-regulated the protein level of NRF-1, PINK1, Parkin, and LC3 II, promoted the recruitment of the cytosolic Parkin, indicating the initiation of the PINK1/Parkin-mediated mitophagy in WRL68 cells. When NRF-1 was down-regulated by NRF-1 specific shRNA, the protein level of PINK1 and Parkin as well as the mitophagy level were significantly decreased. After IH preconditioning, the protein level of PINK1 and the recruitment of Parkin in CCCP-treated group were significantly higher than that of the control group, indicating the increased mitophagy capacity. And the increased mitophagy capacity induced by IH preconditioning was also reduced by down-regulation of NRF-1. Furthermore, the protective effect of IH preconditioning against H2O2-induced oxidative injury in WRL68 cells was inhibited when NRF-1 or PINK1 was down-regulated by specific shRNA. Mitochondrial ROS generation may be responsible for the increased expression of NRF-1 induced by IH preconditioning. In conclusion, the PINK1/Parkin-mediated mitophagy regulated by NRF-1 was involved in IH preconditioning-induced protective effect against oxidative cellular injury in hepatic cells.
Collapse
Affiliation(s)
- Yapeng Lu
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China.
| | - Jiangpei Bian
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Huiwen Kan
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Wangwang Ding
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Dan Wang
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Xueting Wang
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Qianqian Luo
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Xiaomei Wu
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Li Zhu
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China.
| |
Collapse
|
38
|
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
|
39
|
Li H, Luo Y, Liu P, Liu P, Hua W, Zhang Y, Zhang L, Li Z, Xing P, Zhang Y, Hong B, Yang P, Liu J. Exosomes containing miR-451a is involved in the protective effect of cerebral ischemic preconditioning against cerebral ischemia and reperfusion injury. CNS Neurosci Ther 2021; 27:564-576. [PMID: 33533575 PMCID: PMC8025619 DOI: 10.1111/cns.13612] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 12/23/2022] Open
Abstract
Aim To study the role of exosomes in the protective effect of cerebral ischemic preconditioning (cerebral‐IPC) against cerebral I/R injury. Method Mouse models of cerebral‐IPC and MCAO/R were established as described previously, and their behavioral, pathological, and proteomic changes were analyzed. Neuro‐2a subjected to OGD/R were treated with exosomes isolated from the plasma of sham‐operated and cerebral‐IPC mice. The differentially expressed miRNAs between exosomes derived from sham‐operated (S‐exosomes) and preconditioned (IPC‐exosomes) mice were identified through miRNA array, and their targets were identified through database search. The control and OGD/R cells were treated with the IPC‐exosomes, miRNA mimic or target protein inhibitor, and their viability, oxidative, stress and apoptosis rates were measured. The activated pathways were identified by analyzing the levels of relevant proteins. Results Cerebral‐IPC mitigated the cerebral injury following ischemia and reperfusion, and increased the number of plasma exosomes. IPC‐exosomes increased the survival of Neuro‐2a cells after OGD/R. The miR‐451a targeting Rac1 was upregulated in the IPC‐exosomes relative to S‐exosomes. The miR‐451a mimic and the Rac1 inhibitor NSC23766 reversed OGD/R‐mediated activation of Rac1 and its downstream pathways. Conclusion Cerebral‐IPC ameliorated cerebral I/R injury by inducing the release of exosomes containing miR‐451a.
Collapse
Affiliation(s)
- He Li
- Stroke Center, Changhai Hospital, Shanghai, China.,Department of neurosurgery, Changhai Hospital, Shanghai, China
| | - Yin Luo
- Department of neurosurgery, Changhai Hospital, Shanghai, China.,Department of Biomedical Engineering, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Liu
- Stroke Center, Changhai Hospital, Shanghai, China.,Department of neurosurgery, Changhai Hospital, Shanghai, China
| | - Pei Liu
- Stroke Center, Changhai Hospital, Shanghai, China.,Department of neurosurgery, Changhai Hospital, Shanghai, China
| | - Weilong Hua
- Stroke Center, Changhai Hospital, Shanghai, China.,Department of neurosurgery, Changhai Hospital, Shanghai, China
| | - Yongxin Zhang
- Stroke Center, Changhai Hospital, Shanghai, China.,Department of neurosurgery, Changhai Hospital, Shanghai, China
| | - Lei Zhang
- Stroke Center, Changhai Hospital, Shanghai, China
| | - Zifu Li
- Stroke Center, Changhai Hospital, Shanghai, China.,Department of neurosurgery, Changhai Hospital, Shanghai, China
| | - Pengfei Xing
- Stroke Center, Changhai Hospital, Shanghai, China
| | | | - Bo Hong
- Stroke Center, Changhai Hospital, Shanghai, China.,Department of neurosurgery, Changhai Hospital, Shanghai, China
| | - Pengfei Yang
- Stroke Center, Changhai Hospital, Shanghai, China.,Department of neurosurgery, Changhai Hospital, Shanghai, China
| | - Jianmin Liu
- Stroke Center, Changhai Hospital, Shanghai, China.,Department of neurosurgery, Changhai Hospital, Shanghai, China
| |
Collapse
|
40
|
De Lazzari F, Prag HA, Gruszczyk AV, Whitworth AJ, Bisaglia M. DJ-1: A promising therapeutic candidate for ischemia-reperfusion injury. Redox Biol 2021; 41:101884. [PMID: 33561740 PMCID: PMC7872972 DOI: 10.1016/j.redox.2021.101884] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/13/2021] [Accepted: 01/25/2021] [Indexed: 12/31/2022] Open
Abstract
DJ-1 is a multifaceted protein with pleiotropic functions that has been implicated in multiple diseases, ranging from neurodegeneration to cancer and ischemia-reperfusion injury. Ischemia is a complex pathological state arising when tissues and organs do not receive adequate levels of oxygen and nutrients. When the blood flow is restored, significant damage occurs over and above that of ischemia alone and is termed ischemia-reperfusion injury. Despite great efforts in the scientific community to ameliorate this pathology, its complex nature has rendered it challenging to obtain satisfactory treatments that translate to the clinic. In this review, we will describe the recent findings on the participation of the protein DJ-1 in the pathophysiology of ischemia-reperfusion injury, firstly introducing the features and functions of DJ-1 and, successively highlighting the therapeutic potential of the protein. DJ-1 has been shown to confer protection in ischemia-reperfusion injury models. DJ-1 protection relies on the activation of antioxidant signaling pathways. DJ-1 regulates mitochondrial homeostasis during ischemia and reperfusion. DJ-1 seems to modulate ion homeostasis during ischemia and reperfusion. DJ-1 may represent a promising therapeutic target for ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Federica De Lazzari
- Physiology, Genetics and Behaviour Unit, Department of Biology, University of Padova, 35131, Padova, Italy
| | - Hiran A Prag
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Anja V Gruszczyk
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Alexander J Whitworth
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Marco Bisaglia
- Physiology, Genetics and Behaviour Unit, Department of Biology, University of Padova, 35131, Padova, Italy.
| |
Collapse
|
41
|
Incognito AV, Millar PJ, Pyle WG. Remote ischemic conditioning for acute respiratory distress syndrome in COVID-19. Am J Physiol Lung Cell Mol Physiol 2021; 320:L331-L338. [PMID: 33404365 PMCID: PMC7938644 DOI: 10.1152/ajplung.00223.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Acute respiratory distress syndrome and subsequent respiratory failure remains the leading cause of death (>80%) in patients severely impacted by COVID-19. The lack of clinically effective therapies for COVID-19 calls for the consideration of novel adjunct therapeutic approaches. Though novel antiviral treatments and vaccination hold promise in control and prevention of early disease, it is noteworthy that in severe cases of COVID-19, addressing "run-away" inflammatory cascades are likely more relevant for improvement of clinical outcomes. Viral loads may decrease in severe, end-stage coronavirus cases, but a systemically damaging cytokine storm persists and mediates multiple organ injury. Remote ischemic conditioning (RIC) of the limbs has shown potential in recent years to protect the lungs and other organs against pathological conditions similar to that observed in COVID-19. We review the efficacy of RIC in protecting the lungs against acute injury and current points of consideration. The beneficial effects of RIC on lung injury along with other related cardiovascular complications are discussed, as are the limitations presented by sex and aging. This adjunct therapy is highly feasible, noninvasive, and proven to be safe in clinical conditions. If proven effective in clinical trials for acute respiratory distress syndrome and COVID-19, application in the clinical setting could be immediately implemented to improve outcomes.
Collapse
Affiliation(s)
- Anthony V Incognito
- Department of Human Health and Nutritional Sciences, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Philip J Millar
- Department of Human Health and Nutritional Sciences, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada.,Toronto General Research Institute, Toronto, Ontario, Canada
| | - W Glen Pyle
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada.,IMPART Team Canada Investigator Network, Dalhousie Medicine, Dalhousie University, Saint John, New Brunswick, Canada
| |
Collapse
|
42
|
Su Y, Xu R, Zhang R, Qu Y, Zuo W, Ji Z, Geng H, Pan M, Ma G. N6-methyladenosine methyltransferase plays a role in hypoxic preconditioning partially through the interaction with lncRNA H19. Acta Biochim Biophys Sin (Shanghai) 2020; 52:1306-1315. [PMID: 33197240 DOI: 10.1093/abbs/gmaa130] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Indexed: 11/13/2022] Open
Abstract
N6-methyladenosine (m6A), a methylation in the N6 position of adenosine especially in the mRNA, exerts diverse physiological and pathological functions. However, the precise role of m6A methylation in hypoxic preconditioning (HPC) is still unknown. Here, we observed that HPC treatment protected H9c2 cells against H2O2-induced injury, upregulated the m6A level in the total RNA and the expression of methyltransferase like 3 (METTL3), methyltransferase like 14 (METTL14), and long noncoding RNA (lncRNA) H19. Either knockdown of METTL3 or METTL14 notably reversed the HPC-induced enhancement of cell viability, anti-apoptosis ability, and H19 expression. Methylated RNA immunoprecipitation (IP) indicated that knockdown of METTL3 or METTL14 decreased m6A level in the lncRNA H19. Gain-of-function assay demonstrated that H19 overexpression could partially rescue the decreased protection mediated by METTL3 or METTL14 knockdown in HPC-treated H9c2 cells. RNA binding protein immunoprecipitation (RIP) assay showed that METTL3 and METTL14 could directly bind with H19. Our study identified a novel pattern of posttranscriptional regulation in HPC treatment. Since METTL3, METTL14, and lncRNA H19 were involved in HPC protection, they could be considered as potential biomarkers and therapeutic targets in HPC-derived cardiac rehabilitation and therapeutic approaches.
Collapse
Affiliation(s)
- Yamin Su
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Rongfeng Xu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Rui Zhang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yangyang Qu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Wenjie Zuo
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhenjun Ji
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Haihua Geng
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Min Pan
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
43
|
Kooman JP, Stenvinkel P, Shiels PG, Feelisch M, Canaud B, Kotanko P. The oxygen cascade in patients treated with hemodialysis and native high-altitude dwellers: lessons from extreme physiology to benefit patients with end-stage renal disease. Am J Physiol Renal Physiol 2020; 320:F249-F261. [PMID: 33356957 DOI: 10.1152/ajprenal.00540.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Patients treated with hemodialysis (HD) repeatedly undergo intradialytic low arterial oxygen saturation and low central venous oxygen saturation, reflecting an imbalance between upper body systemic oxygen supply and demand, which are associated with increased mortality. Abnormalities along the entire oxygen cascade, with impaired diffusive and convective oxygen transport, contribute to the reduced tissue oxygen supply. HD treatment impairs pulmonary gas exchange and reduces ventilatory drive, whereas ultrafiltration can reduce tissue perfusion due to a decline in cardiac output. In addition to these factors, capillary rarefaction and reduced mitochondrial efficacy can further affect the balance between cellular oxygen supply and demand. Whereas it has been convincingly demonstrated that a reduced perfusion of heart and brain during HD contributes to organ damage, the significance of systemic hypoxia remains uncertain, although it may contribute to oxidative stress, systemic inflammation, and accelerated senescence. These abnormalities along the oxygen cascade of patients treated with HD appear to be diametrically opposite to the situation in Tibetan highlanders and Sherpa, whose physiology adapted to the inescapable hypobaric hypoxia of their living environment over many generations. Their adaptation includes pulmonary, vascular, and metabolic alterations with enhanced capillary density, nitric oxide production, and mitochondrial efficacy without oxidative stress. Improving the tissue oxygen supply in patients treated with HD depends primarily on preventing hemodynamic instability by increasing dialysis time/frequency or prescribing cool dialysis. Whether dietary or pharmacological interventions, such as the administration of L-arginine, fermented food, nitrate, nuclear factor erythroid 2-related factor 2 agonists, or prolyl hydroxylase 2 inhibitors, improve clinical outcome in patients treated with HD warrants future research.
Collapse
Affiliation(s)
- Jeroen P Kooman
- Division of Nephrology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science Technology and Intervention, Karolinska Institutet, Stockholm, Sweden
| | - Paul G Shiels
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Martin Feelisch
- Clinical and Experimental Sciences and Integrative Physiology and Critical Illness Group, Faculty of Medicine, Southampton General Hospital and Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Bernard Canaud
- Montpellier University, School of Medicine, Montpellier, France & Global Medical Office, Fresenius Medical Care, Bad Homburg, Germany
| | - Peter Kotanko
- Renal Research Institute, New York, New York.,Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
44
|
Ren DF, Wang T, Sun GF, Jiang C. Effect of Yiqihuoxue Formula for the treatment of ischemic stroke: A retrospective study. Medicine (Baltimore) 2020; 99:e23260. [PMID: 33285701 PMCID: PMC7717746 DOI: 10.1097/md.0000000000023260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
This retrospective study assessed the feasible effect of Yiqihuoxue Formula (YQHXF) for the treatment of patients with ischemic stroke (IS).A total of 66 patients with IS were included in this retrospective study. All patients received routine treatment, and were divided into two groups: a treatment group (n = 33) and a control group (n = 33). In addition to the routine treatment, all patients in the treatment group also underwent YQHXF treatment. All patients in both groups were treated for a total of 8 weeks. The outcomes were assessed by National Institute of Health Stroke Scale (NIHSS), modified Rankin scale (mRS), Barthel index scale (BIS), stroke-specific quality of life (SS-QOL) scale, and adverse events. All outcomes were measured before and after the treatment.After treatment, patients in the treatment group showed better improvements in NIHSS scale (P = .01), mRS (P < .01), BIS (P = .04), and SS-QOL scale (P = .04), than patients in the control group. No treatment-associated adverse events were recorded in this study.The results of this study indicated that YQHXF may benefit for patients with IS.
Collapse
Affiliation(s)
- Dong-Feng Ren
- Department of Internal Medicine, The First Hospital of Yulin, Yulin, Shaanxi
| | - Ting Wang
- School of Economics and Management, Xidian University, Xi’an
| | - Guo-Fang Sun
- Department of Internal Medicine, The First Hospital of Yulin, Yulin, Shaanxi
| | - Chao Jiang
- The Third Department of Neurology, The Second Affiliated Hospital of Xi’an Medical University, Xi’an, Shaanxi
- Department of Emergency, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
45
|
Jones VM, Suarez-Martinez AD, Hodges NA, Murfee WL, Llull R, Katz AJ. A clinical perspective on adipose-derived cell therapy for enhancing microvascular health and function: Implications and applications for reconstructive surgery. Microcirculation 2020; 28:e12672. [PMID: 33174272 DOI: 10.1111/micc.12672] [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: 06/03/2020] [Revised: 09/18/2020] [Accepted: 11/04/2020] [Indexed: 12/21/2022]
Abstract
Restoration of form and function requires apposition of tissues in the form of flaps to reconstitute local perfusion. Successful reconstruction relies on flap survival and its integration with the recipient bed. The flap's precariously perfused hypoxic areas undergo adaptive microvascular changes both internally and in connection with the recipient bed. A cell-mediated, coordinated response to hypoxia drives these adaptive processes, restoring a tissue's normoxic homeostasis via de novo vasculogenesis, sprouting angiogenesis, and stabilizing arterialization. As cells exert prolonged and coordinated effects on site, their use as biological agents merit translational consideration of sourcing angio-competent cells and delivering them to territories enduring microcirculatory acclimatization. Angio-competent cells abound in adipose tissue: a reliable, accessible, and expendable source of adipose-derived cells (ADC). When subject to enzymatic digestion and centrifugation, adipose tissue separates its various ADC: A subset of buoyant oil-dense adipocytes (the tissue's parenchymal component) accumulates on a supra-natant layer, whereas the mesenchymal component remains in the infra-natant sediment, containing the tissue's stromal vascular fraction (SVF), where angio-component cells abound. The SVF can be further manipulated, selected, or culture expanded into more specific stromal subsets (herein defined as adipose stromal cells, ASC). While promising clinical applications for ADC await clinical proof and regulatory authorization, basic science investigation is needed to elucidate the specific ADC mechanisms that influence microvascular growth, remodeling, and function following flap surgery. The objective of this article is to share the clinical perspectives of reconstructive plastic surgeons regarding the use of ADC-based therapies to help with flap tissue integration, revascularization, and wound healing. Specifically, the focus will be on considering the potential for ADC as therapeutic agents and how their clinical application motivates basic science opportunities.
Collapse
Affiliation(s)
- V Morgan Jones
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Ariana D Suarez-Martinez
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Nicholas A Hodges
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Walter L Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Ramon Llull
- Department of Plastic Surgery, Hospital Quiron Salud PalmaPlanas, Palma, Spain
| | - Adam J Katz
- Department of Plastic and Reconstructive Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
46
|
Thorne AM, Huang H, O'Brien DP, Eijken M, Krogstrup NV, Norregaard R, Møller B, Ploeg RJ, Jespersen B, Kessler BM. Subclinical effects of remote ischaemic conditioning in human kidney transplants revealed by quantitative proteomics. Clin Proteomics 2020; 17:39. [PMID: 33292164 PMCID: PMC7607690 DOI: 10.1186/s12014-020-09301-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 10/10/2020] [Indexed: 01/13/2023] Open
Abstract
Background Remote ischaemic conditioning (RIC) is currently being explored as a non-invasive method to attenuate ischaemia/reperfusion injuries in organs. A randomised clinical study (CONTEXT) evaluated the effects of RIC compared to non-RIC controls in human kidney transplants. Methods RIC was induced prior to kidney reperfusion by episodes of obstruction to arterial flow in the leg opposite the transplant using a tourniquet (4 × 5 min). Although RIC did not lead to clinical improvement of transplant outcomes, we explored whether RIC induced molecular changes through precision analysis of CONTEXT recipient plasma and kidney tissue samples by high-resolution tandem mass spectrometry (MS/MS). Results We observed an accumulation of muscle derived proteins and altered amino acid metabolism in kidney tissue proteomes, likely provoked by RIC, which was not reflected in plasma. In addition, MS/MS analysis demonstrated transient upregulation of several acute phase response proteins (SAA1, SAA2, CRP) in plasma, 1 and 5 days post-transplant in RIC and non-RIC conditions with a variable effect on the magnitude of acute inflammation. Conclusions Together, our results indicate sub-clinical systemic and organ-localised effects of RIC.
Collapse
Affiliation(s)
- Adam M Thorne
- Nuffield Department of Surgical Sciences and Oxford Biomedical Research Centre, University of Oxford, Oxford, UK.,Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Honglei Huang
- Nuffield Department of Surgical Sciences and Oxford Biomedical Research Centre, University of Oxford, Oxford, UK.,Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Darragh P O'Brien
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Marco Eijken
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Nicoline Valentina Krogstrup
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark.,Department of Renal Medicine, Rigshospitalet, Copenhagen, Denmark
| | - Rikke Norregaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Bjarne Møller
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Rutger J Ploeg
- Nuffield Department of Surgical Sciences and Oxford Biomedical Research Centre, University of Oxford, Oxford, UK.
| | - Bente Jespersen
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark. .,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Benedikt M Kessler
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
47
|
Bratek E, Ziembowicz A, Salinska E. N-Acetylaspartylglutamate (NAAG) Pretreatment Reduces Hypoxic-Ischemic Brain Damage and Oxidative Stress in Neonatal Rats. Antioxidants (Basel) 2020; 9:antiox9090877. [PMID: 32957477 PMCID: PMC7555246 DOI: 10.3390/antiox9090877] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022] Open
Abstract
N-acetylaspartylglutamate (NAAG), the most abundant peptide transmitter in the mammalian nervous system, activates mGluR3 at presynaptic sites, inhibiting the release of glutamate, and acts on mGluR3 on astrocytes, stimulating the release of neuroprotective growth factors (TGF-β). NAAG can also affect N-methyl-d-aspartate (NMDA) receptors in both synaptic and extrasynaptic regions. NAAG reduces neurodegeneration in a neonatal rat model of hypoxia-ischemia (HI), although the exact mechanism is not fully recognized. In the present study, the effect of NAAG application 24 or 1 h before experimental birth asphyxia on oxidative stress markers and the potential mechanisms of neuroprotection on 7-day old rats was investigated. The intraperitoneal application of NAAG at either time point before HI significantly reduced the weight deficit of the ischemic brain hemisphere, radical oxygen species (ROS) content and activity of antioxidant enzymes, and increased the concentration of reduced glutathione (GSH). No additional increase in the TGF-β concentration was observed after NAAG application. The fast metabolism of NAAG and the decrease in TGF-β concentration that resulted from NAAG pretreatment, performed up to 24 h before HI, excluded the involvement mGluR3 in neuroprotection. The observed effect may be explained by the activation of NMDA receptors induced by NAAG pretreatment 24 h before HI. Inhibition of the NAAG effect by memantine supports this conclusion. NAAG preconditioning 1 h before HI results in a mixture of mGluR3 and NMDA receptor activation. Preconditioning with NAAG induces the antioxidative defense system triggered by mild excitotoxicity in neurons. Moreover, this response to NAAG pretreatment is consistent with the commonly accepted mechanism of preconditioning. However, this theory requires further investigation.
Collapse
|
48
|
Chacaroun S, Borowik A, Doutreleau S, Belaidi E, Wuyam B, Tamisier R, Pépin JL, Flore P, Verges S. Cardiovascular and metabolic responses to passive hypoxic conditioning in overweight and mildly obese individuals. Am J Physiol Regul Integr Comp Physiol 2020; 319:R211-R222. [PMID: 32609532 DOI: 10.1152/ajpregu.00311.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Although severe intermittent hypoxia (IH) is well known to induce deleterious cardiometabolic consequences, moderate IH may induce positive effects in obese individuals. The present study aimed to evaluate the effect of two hypoxic conditioning programs on cardiovascular and metabolic health status of overweight or obese individuals. In this randomized single-blind controlled study, 35 subjects (54 ± 9.3 yr, 31.7 ± 3.5 kg/m2) were randomized into three 8-wk interventions (three 1-h sessions per week): sustained hypoxia (SH), arterial oxygen saturation ([Formula: see text]) = 75%; IH, 5 min [Formula: see text] = 75% - 3 min normoxia; normoxia. Ventilation, heart rate, blood pressure, and tissue oxygenation were measured during the first and last hypoxic conditioning sessions. Vascular function, blood glucose and insulin, lipid profile, nitric oxide metabolites, and oxidative stress were evaluated before and after the interventions. Both SH and IH increased ventilation in hypoxia (+1.8 ± 2.1 and +2.3 ± 3.6 L/min, respectively; P < 0.05) and reduced normoxic diastolic blood pressure (-12 ± 15 and -13 ± 10 mmHg, respectively; P < 0.05), whereas changes in normoxic systolic blood pressure were not significant (+3 ± 9 and -6 ± 13 mmHg, respectively; P > 0.05). IH only reduced heart rate variability (e.g., root-mean-square difference of successive normal R-R intervals in normoxia -21 ± 35%; P < 0.05). Both SH and IH induced no significant change in body mass index, vascular function, blood glucose, insulin and lipid profile, nitric oxide metabolites, or oxidative stress, except for an increase in superoxide dismutase activity following SH. This study indicates that passive hypoxic conditioning in obese individuals induces some positive cardiovascular and respiratory improvements despite no change in anthropometric data and even a reduction in heart rate variability during IH exposure.
Collapse
Affiliation(s)
- Samarmar Chacaroun
- HP2 laboratory, Univiversité Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
| | - Anna Borowik
- HP2 laboratory, Univiversité Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
| | - Stephane Doutreleau
- HP2 laboratory, Univiversité Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
| | - Elise Belaidi
- HP2 laboratory, Univiversité Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
| | - Bernard Wuyam
- HP2 laboratory, Univiversité Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
| | - Renaud Tamisier
- HP2 laboratory, Univiversité Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
| | - Jean-Louis Pépin
- HP2 laboratory, Univiversité Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
| | - Patrice Flore
- HP2 laboratory, Univiversité Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
| | - Samuel Verges
- HP2 laboratory, Univiversité Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
| |
Collapse
|
49
|
Yan G, Zhao H, Hong X. LncRNA MACC1-AS1 attenuates microvascular endothelial cell injury and promotes angiogenesis under hypoxic conditions via modulating miR-6867-5p/TWIST1 in human brain microvascular endothelial cells. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:876. [PMID: 32793720 DOI: 10.21037/atm-20-4915] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Hypoxia following ischemic stroke is a common cause of brain insults. Mounting evidence suggests that long non-coding RNAs (lncRNAs) play a vital role in regulating certain physiological and pathological processes including ischemic stroke. For the first time, the present study investigated the effects and mechanism of LncRNA MACC1-AS1 on hypoxia-induced human brain microvascular endothelial cells (HBMECs). Methods LncRNA MACC1-AS1 levels in HBMECs were detected via reverse transcription quantitative polymerase chain reaction (RT-qPCR) assay. Reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD), and catalase (CAT), were detected using their respective kits. Flow cytometry and clone formation assay were performed to evaluate the effects of lncRNA MACC1-AS1 on cell apoptosis and cell proliferation respectively. Angiogenesis capacity was evaluated via tube formation assay. Transwell migration assay was performed for assessment of cell migration, Western blot assay was performed for measurement of Twist1 and VE-cadherin level, and permeability assay was performed for evaluation of the cell barrier function. The target gene was predicted via bioinformatics online tool and validated through luciferase reporter assay and RNA pull-down assay. Results LncRNA MACC1-AS1 was downregulated in hypoxia-induced HBMECs. Overexpression of LncRNA MACC1-AS1 reduced cell apoptosis and oxidative stress, while promoting cell proliferation, migration, and angiogenesis. Moreover, LncRNA MACC1-AS1 overexpression reduced cell permeability and elevated VE-cadherin level, which contributed to maintaining cell barrier function. TWIST1 was validated as the target of miR-6867-5p which was further targeted by lncRNA MACC1-AS1. Thus, LncRNA MACC1-AS1 functions in hypoxia-induced HBMECs by regulating miR-6867-5p/TWIST1. Conclusions In this study, we found that LncRNA MACC1-AS1 exerted a protective role in hypoxia-induced HBMECs via regulating miR-6867-5p/TWIST1, indicating a new therapeutic strategy for future ischemic stroke therapy.
Collapse
Affiliation(s)
- Guangjun Yan
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Haomin Zhao
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xin Hong
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
50
|
Zhao Y, Zhou Y, Ma X, Liu X, Zhao Y, Liu X. DDAH-1 via HIF-1 target genes improves cerebral ischemic tolerance after hypoxic preconditioning and middle cerebral artery occlusion-reperfusion. Nitric Oxide 2020; 95:17-28. [DOI: 10.1016/j.niox.2019.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/04/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023]
|