1
|
Darwish D, Kumar P, Urs K, Dave S. Inhaled Anesthetics: Beyond the Operating Room. J Clin Med 2024; 13:7513. [PMID: 39768435 PMCID: PMC11679802 DOI: 10.3390/jcm13247513] [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: 11/19/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
The development of inhaled anesthetics (IAs) has a rich history dating back many centuries. In modern times they have played a pivotal role in anesthesia and critical care by allowing deep sedation during periods of critical illness and surgery. In addition to their sedating effects, they have many systemic effects allowing for therapy beyond surgical anesthesia. In this narrative review we chronicle the evolution of IAs, from early volatile agents such as ether to the contemporary use of halogenated hydrocarbons. This is followed by a discussion of the mechanisms of action of these agents which primarily involve the modulation of lipid membrane properties and ion channel activity. IAs' systemic effects are also examined, including their effects on the cardiovascular, respiratory, hepatic, renal and nervous systems. We discuss of the role of IAs in treating systemic disease processes including ischemic stroke, delayed cerebral ischemia, status epilepticus, status asthmaticus, myocardial ischemia, and intensive care sedation. We conclude with a review of the practical and logistical challenges of utilizing IAs outside the operating room as well as directions for future research. This review highlights the expanding clinical utility of IAs and their evolving role in the management of a diverse range of disease processes, offering new avenues for therapeutic exploration beyond anesthesia.
Collapse
Affiliation(s)
- Dana Darwish
- Department of Anesthesiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Pooja Kumar
- School of Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Khushi Urs
- University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Siddharth Dave
- Department of Anesthesiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
2
|
Farkas E, Rose CR. A dangerous liaison: Spreading depolarization and tissue acidification in cerebral ischemia. J Cereb Blood Flow Metab 2024:271678X241289756. [PMID: 39535276 DOI: 10.1177/0271678x241289756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Brain pH is precisely regulated, and pH transients associated with activity are rapidly restored under physiological conditions. During ischemia, the brain's ability to buffer pH changes is rapidly depleted. Tissue oxygen deprivation causes a shift from aerobic to anaerobic metabolism and the accumulation of lactic acid and protons. Although the degree of tissue acidosis resulting from ischemia depends on the severity of the ischemia, spreading depolarization (SD) events emerge as central elements to determining ischemic tissue acidosis. A marked decrease in tissue pH during cerebral ischemia may exacerbate neuronal injury, which has become known as acidotoxicity, in analogy to excitotoxicity. The cellular pathways underlying acidotoxicity have recently been described in increasing detail. The molecular structure of acid or base carriers and acidosis-activated ion channels, the precise (dys)homeostatic conditions under which they are activated, and their possible role in severe ischemia have been addressed. The expanded understanding of acidotoxic mechanisms now provides an opportunity to reevaluate the contexts that lead to acidotoxic injury. Here, we review the specific cellular pathways of acidotoxicity and demonstrate that SD plays a central role in activating the molecular machinery leading to acid-induced damage. We propose that SD is a key contributor to acidotoxic injury in cerebral ischemia.
Collapse
Affiliation(s)
- Eszter Farkas
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Christine R Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
3
|
Zheng H, Xiao X, Han Y, Wang P, Zang L, Wang L, Zhao Y, Shi P, Yang P, Guo C, Xue J, Zhao X. Research progress of propofol in alleviating cerebral ischemia/reperfusion injury. Pharmacol Rep 2024; 76:962-980. [PMID: 38954373 DOI: 10.1007/s43440-024-00620-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/21/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024]
Abstract
Ischemic stroke is a leading cause of adult disability and death worldwide. The primary treatment for cerebral ischemia patients is to restore blood supply to the ischemic region as quickly as possible. However, in most cases, more severe tissue damage occurs, which is known as cerebral ischemia/reperfusion (I/R) injury. The pathological mechanisms of brain I/R injury include mitochondrial dysfunction, oxidative stress, excitotoxicity, calcium overload, neuroinflammation, programmed cell death and others. Propofol (2,6-diisopropylphenol), a short-acting intravenous anesthetic, possesses not only sedative and hypnotic effects but also immunomodulatory and neuroprotective effects. Numerous studies have reported the protective properties of propofol during brain I/R injury. In this review, we summarize the potential protective mechanisms of propofol to provide insights for its better clinical application in alleviating cerebral I/R injury.
Collapse
Affiliation(s)
- Haijing Zheng
- Basic Medical College, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
- Zhengzhou Central Hospital, Zhengzhou, China
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Xian Xiao
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Yiming Han
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Pengwei Wang
- Department of Pharmacy, the First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Weihui, Henan, 453100, China
| | - Lili Zang
- Department of Surgery, the First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Weihui, China
| | - Lilin Wang
- Department of Pediatric Surgery, the First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Weihui, China
| | - Yinuo Zhao
- Basic Medical College, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Peijie Shi
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Pengfei Yang
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China.
| | - Chao Guo
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China.
| | - Jintao Xue
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China.
| | - Xinghua Zhao
- Basic Medical College, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China.
| |
Collapse
|
4
|
Miyara SJ, Shinozaki K, Hayashida K, Shoaib M, Choudhary RC, Zafeiropoulos S, Guevara S, Kim J, Molmenti EP, Volpe BT, Becker LB. Differential Mitochondrial Bioenergetics in Neurons and Astrocytes Following Ischemia-Reperfusion Injury and Hypothermia. Biomedicines 2024; 12:1705. [PMID: 39200170 PMCID: PMC11352110 DOI: 10.3390/biomedicines12081705] [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: 03/11/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 09/02/2024] Open
Abstract
The close interaction between neurons and astrocytes has been extensively studied. However, the specific behavior of these cells after ischemia-reperfusion injury and hypothermia remains poorly characterized. A growing body of evidence suggests that mitochondria function and putative transference between neurons and astrocytes may play a fundamental role in adaptive and homeostatic responses after systemic insults such as cardiac arrest, which highlights the importance of a better understanding of how neurons and astrocytes behave individually in these settings. Brain injury is one of the most important challenges in post-cardiac arrest syndrome, and therapeutic hypothermia remains the single, gold standard treatment for neuroprotection after cardiac arrest. In our study, we modeled ischemia-reperfusion injury by using in vitro enhanced oxygen-glucose deprivation and reperfusion (eOGD-R) and subsequent hypothermia (HPT) (31.5 °C) to cell lines of neurons (HT-22) and astrocytes (C8-D1A) with/without hypothermia. Using cell lysis (LDH; lactate dehydrogenase) as a measure of membrane integrity and cell viability, we found that neurons were more susceptible to eOGD-R when compared with astrocytes. However, they benefited significantly from HPT, while the HPT effect after eOGD-R on astrocytes was negligible. Similarly, eOGD-R caused a more significant reduction in adenosine triphosphate (ATP) in neurons than astrocytes, and the ATP-enhancing effects from HPT were more prominent in neurons than astrocytes. In both neurons and astrocytes, measurement of reactive oxygen species (ROS) revealed higher ROS output following eOGD-R, with a non-significant trend of differential reduction observed in neurons. HPT after eOGD-R effectively downregulated ROS in both cells; however, the effect was significantly more effective in neurons. Lipid peroxidation was higher after eOGD-R in neurons, while in astrocytes, the increase was not statistically significant. Interestingly, HPT had similar effects on the reduction in lipoperoxidation after eOGD-R with both types of cells. While glutathione (GSH) levels were downregulated after eOGD-R in both cells, HPT enhanced GSH in astrocytes, but worsened GSH in neurons. In conclusion, neuron and astrocyte cultures respond differently to eOGD-R and eOGD-R + HTP treatments. Neurons showed higher sensitivity to ischemia-reperfusion insults than astrocytes; however, they benefited more from HPT therapy. These data suggest that given the differential effects from HPT in neurons and astrocytes, future therapeutic developments could potentially enhance HPT outcomes by means of neuronal and astrocytic targeted therapies.
Collapse
Affiliation(s)
- Santiago J. Miyara
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY 11030, USA
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Koichiro Shinozaki
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Emergency Medicine, Northwell Health, Manhasset, NY 11030, USA
| | - Kei Hayashida
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Emergency Medicine, Northwell Health, Manhasset, NY 11030, USA
| | - Muhammad Shoaib
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | | | | | - Sara Guevara
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Junhwan Kim
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Ernesto P. Molmenti
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Surgery, Renown Health, Reno, NV 89502, USA
| | - Bruce T. Volpe
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY 11030, USA
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Lance B. Becker
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY 11030, USA
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Emergency Medicine, Northwell Health, Manhasset, NY 11030, USA
- Department of Emergency Medicine, Kindai University Faculty of Medicine, Osaka 589-8511, Japan
| |
Collapse
|
5
|
Kumar Saini S, Singh D. Mitochondrial mechanisms in Cerebral Ischemia-Reperfusion Injury: Unravelling the intricacies. Mitochondrion 2024; 77:101883. [PMID: 38631511 DOI: 10.1016/j.mito.2024.101883] [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: 01/01/2024] [Revised: 04/05/2024] [Accepted: 04/13/2024] [Indexed: 04/19/2024]
Abstract
Cerebral ischemic stroke is a major contributor to physical impairments and premature death worldwide. The available reperfusion therapies for stroke in the form of mechanical thrombectomy and intravenous thrombolysis increase the risk of cerebral ischemia-reperfusion (I-R) injury due to sudden restoration of blood supply to the ischemic region. The injury is manifested by hemorrhagic transformation, worsening of neurological impairments, cerebral edema, and progression to infarction in surviving patients. A complex network of multiple pathological processes has been known to be involved in the pathogenesis of I-R injury. Primarily, 3 major contributors namely oxidative stress, neuroinflammation, and mitochondrial failure have been well studied in I-R injury. A transcription factor, Nrf2 (Nuclear factor erythroid 2-related factor 2) plays a crucial defensive role in resisting the deleterious effects of I-R injury and potentiating the cellular protective mechanisms. In this review, we delve into the critical function of mitochondria and Nrf2 in the context of cerebral I-R injury. We summarized how oxidative stress, neuroinflammation, and mitochondrial anomaly contribute to the pathophysiology of I-R injury and further elaborated the role of Nrf2 as a pivotal guardian of cellular integrity. The review further highlighted Nrf2 as a putative therapeutic target for mitochondrial dysfunction in cerebral I-R injury management.
Collapse
Affiliation(s)
- Shiv Kumar Saini
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Damanpreet Singh
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
6
|
Ning W, Li L, Wang R, Zhang B, Yang S, Zhang L, Fan X, Shen Y, Zhang Y, Zhao M, Wang Y, Liang P, Wang S. Electroacupuncture pretreatment enhances the calcium efflux activity of Na +/Ca 2+ exchanger to attenuate cerebral injury by PI3K/Akt-mediated NCX1 upregulation after focal cerebral ischaemia. Heliyon 2024; 10:e33265. [PMID: 39022107 PMCID: PMC11253542 DOI: 10.1016/j.heliyon.2024.e33265] [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: 12/20/2023] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024] Open
Abstract
Electroacupuncture pretreatment is considered as an optimal strategy for inducing cerebral ischaemic tolerance. However, the underlying neuroprotective mechanism of this approach has never been explored from the perspective of calcium homeostasis. Intracellular calcium overload is a key inducer of cascade neuronal injury in the early stage after cerebral ischaemia attack and the Na+/Ca2+ exchanger (NCX) is the main plasma membrane calcium extrusion pathway maintaining post-ischaemic calcium homeostasis. This study aims to investigate whether the regulation of NCX-mediated calcium transport contributes to the cerebroprotective effect of electroacupuncture pretreatment against ischaemic injury and to elucidate the underlying mechanisms involved in this process. Following five days of repeated electroacupuncture stimulation on Baihui (GV20), Neiguan (PC6), and Sanyinjiao (SP6) acupoints in rats, in vivo and in vitro models of cerebral ischaemia were induced through middle cerebral artery occlusion and oxygen/glucose deprivation (OGD), respectively. Firstly, we verified the neuroprotective effect of electroacupuncture pretreatment from the perspective of neurological score, infarct volume and neuronal apoptosis. Our findings from brain slice patch-clamp indicated that electroacupuncture pretreatment enhanced the Ca2+ efflux capacity of NCX after OGD. NCX1 expression in the ischaemic penumbra exhibited a consistent decline from 1 to 24 h in MCAO rats. Electroacupuncture pretreatment upregulated the expression of NCX1, especially at 24 h, and silencing NCX1 by short hairpin RNA (shRNA) administration reversed the protective effect of electroacupuncture pretreatment against cerebral ischaemic injury. Furthermore, we administered LY294002, a phosphatidylinositol 3 kinase (PI3K) inhibitor, prior to inducing ischaemia to investigate the upstream regulatory mechanism of electroacupuncture pretreatment on NCX1 expression. Electroacupuncture pretreatment activates PI3K/Akt pathway, leading to an increase in the expression of NCX1, which facilitates calcium extrusion and exerts a neuroprotective effect against cerebral ischaemia. These findings provided a novel insight into the prevention of ischemic stroke and other similar conditions characterized by brain ischaemia or hypoperfusion.
Collapse
Affiliation(s)
- Wenhua Ning
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, China
| | - Li Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin Key Laboratory of Acupuncture and Moxibustion, Tianjin, China
| | - Ruiqi Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Baoyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Sha Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Lili Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xiaonong Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Laboratory of Dosage-Effect Relationship, State Administration of Traditional Chinese Medicine (Level 3), Tianjin, China
| | - Yan Shen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yanan Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Mengxiong Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Peizhe Liang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shu Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
- Key Laboratory of Cerebropathy Acupuncture Therapy of State Administration of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
7
|
Mushtaq U. EP1 receptor: Devil in emperors coat. J Cell Biochem 2023; 124:1105-1114. [PMID: 37450673 DOI: 10.1002/jcb.30436] [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: 01/23/2023] [Revised: 05/20/2023] [Accepted: 06/06/2023] [Indexed: 07/18/2023]
Abstract
EP1 receptor belongs to prostanoid receptors and is activated by prostaglandin E2. The receptor performs contrasting functions in central nervous system (CNS) and other tissues. Although the receptor is neurotoxic and proapoptotic in CNS, it has also been reported to act in an antiapoptotic manner by modulating cell survival, proliferation, invasion, and migration in different types of cancers. The receptor mediates its neurotoxic effects by increasing cytosolic Ca2+ levels, leading to the activation of its downstream target, protein kinase C, in different neurological disorders including Alzheimer's disease, Parkinson's disease, stroke, amyotrophic lateral sclerosis, and epilepsy. Antagonists ONO-8713, SC51089, and SC51322 against EP1 receptor ameliorate the neurotoxic effect by attenuating the neuroinflammation. The receptor also shows increased expression in different types of cancers and has been found to activate different signaling pathways, which lead to the development, progression, and metastasis of different cancers. The receptor stimulates the cell survival pathway by phosphorylating the AKT and PTEN (phosphatase and tensin homolog deleted on chromosome 10) signaling pathways. Although there are limited studies about this receptor and not a single clinical trial has been targeting the EP1 receptor for different neurological disorders or cancer, the receptor is appearing as a potential candidate for therapeutic targets. The aim of this article is to review the recent progress in understanding the pathogenic roles of EP1 receptors in different pathological conditions.
Collapse
Affiliation(s)
- Umar Mushtaq
- Department of Biotechnology, Central University of Kashmir, Ganderbal, India
| |
Collapse
|
8
|
Preziuso A, Piccirillo S, Cerqueni G, Serfilippi T, Terenzi V, Vinciguerra A, Orciani M, Amoroso S, Magi S, Lariccia V. Exploring the Role of NCX1 and NCX3 in an In Vitro Model of Metabolism Impairment: Potential Neuroprotective Targets for Alzheimer's Disease. BIOLOGY 2023; 12:1005. [PMID: 37508434 PMCID: PMC10376230 DOI: 10.3390/biology12071005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023]
Abstract
Alzheimer's disease (AD) is a widespread neurodegenerative disorder, affecting a large number of elderly individuals worldwide. Mitochondrial dysfunction, metabolic alterations, and oxidative stress are regarded as cooperating drivers of the progression of AD. In particular, metabolic impairment amplifies the production of reactive oxygen species (ROS), resulting in detrimental alterations to intracellular Ca2+ regulatory processes. The Na+/Ca2+ exchanger (NCX) proteins are key pathophysiological determinants of Ca2+ and Na+ homeostasis, operating at both the plasma membrane and mitochondria levels. Our study aimed to explore the role of NCX1 and NCX3 in retinoic acid (RA) differentiated SH-SY5Y cells treated with glyceraldehyde (GA), to induce impairment of the default glucose metabolism that typically precedes Aβ deposition or Tau protein phosphorylation in AD. By using an RNA interference-mediated approach to silence either NCX1 or NCX3 expression, we found that, in GA-treated cells, the knocking-down of NCX3 ameliorated cell viability, increased the intracellular ATP production, and reduced the oxidative damage. Remarkably, NCX3 silencing also prevented the enhancement of Aβ and pTau levels and normalized the GA-induced decrease in NCX reverse-mode activity. By contrast, the knocking-down of NCX1 was totally ineffective in preventing GA-induced cytotoxicity except for the increase in ATP synthesis. These findings indicate that NCX3 and NCX1 may differently influence the evolution of AD pathology fostered by glucose metabolic dysfunction, thus providing a potential target for preventing AD.
Collapse
Affiliation(s)
- Alessandra Preziuso
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Silvia Piccirillo
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Giorgia Cerqueni
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Tiziano Serfilippi
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Valentina Terenzi
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Antonio Vinciguerra
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Monia Orciani
- Department of Clinical and Molecular Sciences-Histology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Salvatore Amoroso
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Simona Magi
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Vincenzo Lariccia
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| |
Collapse
|
9
|
Jia J, Deng J, Jin H, Yang J, Nan D, Yu Z, Yu W, Shen Z, Lu Y, Liu R, Wang Z, Qu X, Qiu D, Yang Z, Huang Y. Effect of Dl-3-n-butylphthalide on mitochondrial Cox7c in models of cerebral ischemia/reperfusion injury. Front Pharmacol 2023; 14:1084564. [PMID: 36909178 PMCID: PMC9992206 DOI: 10.3389/fphar.2023.1084564] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 02/07/2023] [Indexed: 02/24/2023] Open
Abstract
Several studies have demonstrated the protective effect of dl-3-n-Butylphthalide (NBP) against cerebral ischemia, which may be related to the attenuation of mitochondrial dysfunction. However, the specific mechanism and targets of NBP in cerebral ischemia/reperfusion remains unclear. In this study, we used a chemical proteomics approach to search for targets of NBP and identified cytochrome C oxidase 7c (Cox7c) as a key interacting target of NBP. Our findings indicated that NBP inhibits mitochondrial apoptosis and reactive oxygen species (ROS) release and increases ATP production through upregulation of Cox7c. Subsequently, mitochondrial respiratory capacity was improved and the HIF-1α/VEGF pathway was upregulated, which contributed to the maintenance of mitochondrial membrane potential and blood brain barrier integrity and promoting angiogenesis. Therefore, our findings provided a novel insight into the mechanisms underlying the neuroprotective effects of NBP, and also proposed for the first time that Cox7c exerts a critical role by protecting mitochondrial function.
Collapse
Affiliation(s)
- Jingjing Jia
- Department of Neurology, Peking University First Hospital, Beijing, China.,National Center for Children's Health, Department of Neurology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Jianwen Deng
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Haiqiang Jin
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Jie Yang
- Leewe Biopharmaceutical Co., Ltd, Xianlin University, Nanjing, China
| | - Ding Nan
- Department of Neurology, Peking University First Hospital, Beijing, China.,Department of Hyperbaric Oxygen, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Zemou Yu
- Department of Neurology, Peking University First Hospital, Beijing, China.,National Center for Children's Health, Department of Neurology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Weiwei Yu
- Department of Neurology, Peking University First Hospital, Beijing, China.,Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Zhiyuan Shen
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Yuxuan Lu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Ran Liu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Xiaozhong Qu
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Dong Qiu
- Beijing National Laboratory for Molecular Sciences (BNLMS), Laboratory of Polymer Physics and Chemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Zhenzhong Yang
- Institute of Polymer Science and Engineering, Department of Chemical Engineering, Tsinghua University, Beijing, China
| | - Yining Huang
- Department of Neurology, Peking University First Hospital, Beijing, China
| |
Collapse
|
10
|
Ballarini E, Malacrida A, Rodriguez-Menendez V, Pozzi E, Canta A, Chiorazzi A, Monza L, Semperboni S, Meregalli C, Carozzi VA, Hashemi M, Nicolini G, Scuteri A, Housley SN, Cavaletti G, Alberti P. Sodium-Calcium Exchanger 2: A Pivotal Role in Oxaliplatin Induced Peripheral Neurotoxicity and Axonal Damage? Int J Mol Sci 2022; 23:10063. [PMID: 36077454 PMCID: PMC9456447 DOI: 10.3390/ijms231710063] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 12/14/2022] Open
Abstract
Oxaliplatin (OHP)-induced peripheral neurotoxicity (OIPN) is a frequent adverse event of colorectal cancer treatment. OIPN encompasses a chronic and an acute syndrome. The latter consists of transient axonal hyperexcitability, due to unbalance in Na+ voltage-operated channels (Na+VOC). This leads to sustained depolarisation which can activate the reverse mode of the Na+/Ca2+ exchanger 2 (NCX2), resulting in toxic Ca2+ accumulation and axonal damage (ADa). We explored the role of NCX2 in in vitro and in vivo settings. Embryonic rat Dorsal Root Ganglia (DRG) organotypic cultures treated with SEA0400 (SEA), a NCX inhibitor, were used to assess neuroprotection in a proof-of-concept and pilot study to exploit NCX modulation to prevent ADa. In vivo, OHP treated mice (7 mg/Kg, i.v., once a week for 8 weeks) were compared with a vehicle-treated group (n = 12 each). Neurophysiological and behavioural testing were performed to characterise acute and chronic OIPN, and morphological analyses were performed to detect ADa. Immunohistochemistry, immunofluorescence, and western blotting (WB) analyses were also performed to demonstrate changes in NCX2 immunoreactivity and protein expression. In vitro, NCX inhibition was matched by ADa mitigation. In the in vivo part, after verifyingboth acute and chronic OIPN had ensued, we confirmed via immunohistochemistry, immunofluorescence, and WB that a significant NCX2 alteration had ensued in the OHP group. Our data suggest NCX2 involvement in ADa development, paving the way to a new line of research to prevent OIPN.
Collapse
Affiliation(s)
- Elisa Ballarini
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Alessio Malacrida
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Virginia Rodriguez-Menendez
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Eleonora Pozzi
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Annalisa Canta
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Alessia Chiorazzi
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Laura Monza
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Sara Semperboni
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Cristina Meregalli
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Valentina Alda Carozzi
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Maryamsadat Hashemi
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Gabriella Nicolini
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Arianna Scuteri
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Stephen N. Housley
- Integrated Cancer Research Center, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Guido Cavaletti
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Paola Alberti
- School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| |
Collapse
|
11
|
Tripathi R, Gupta R, Sahu M, Srivastava D, Das A, Ambasta RK, Kumar P. Free radical biology in neurological manifestations: mechanisms to therapeutics interventions. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:62160-62207. [PMID: 34617231 DOI: 10.1007/s11356-021-16693-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 09/20/2021] [Indexed: 06/13/2023]
Abstract
Recent advancements and growing attention about free radicals (ROS) and redox signaling enable the scientific fraternity to consider their involvement in the pathophysiology of inflammatory diseases, metabolic disorders, and neurological defects. Free radicals increase the concentration of reactive oxygen and nitrogen species in the biological system through different endogenous sources and thus increased the overall oxidative stress. An increase in oxidative stress causes cell death through different signaling mechanisms such as mitochondrial impairment, cell-cycle arrest, DNA damage response, inflammation, negative regulation of protein, and lipid peroxidation. Thus, an appropriate balance between free radicals and antioxidants becomes crucial to maintain physiological function. Since the 1brain requires high oxygen for its functioning, it is highly vulnerable to free radical generation and enhanced ROS in the brain adversely affects axonal regeneration and synaptic plasticity, which results in neuronal cell death. In addition, increased ROS in the brain alters various signaling pathways such as apoptosis, autophagy, inflammation and microglial activation, DNA damage response, and cell-cycle arrest, leading to memory and learning defects. Mounting evidence suggests the potential involvement of micro-RNAs, circular-RNAs, natural and dietary compounds, synthetic inhibitors, and heat-shock proteins as therapeutic agents to combat neurological diseases. Herein, we explain the mechanism of free radical generation and its role in mitochondrial, protein, and lipid peroxidation biology. Further, we discuss the negative role of free radicals in synaptic plasticity and axonal regeneration through the modulation of various signaling molecules and also in the involvement of free radicals in various neurological diseases and their potential therapeutic approaches. The primary cause of free radical generation is drug overdosing, industrial air pollution, toxic heavy metals, ionizing radiation, smoking, alcohol, pesticides, and ultraviolet radiation. Excessive generation of free radicals inside the cell R1Q1 increases reactive oxygen and nitrogen species, which causes oxidative damage. An increase in oxidative damage alters different cellular pathways and processes such as mitochondrial impairment, DNA damage response, cell cycle arrest, and inflammatory response, leading to pathogenesis and progression of neurodegenerative disease other neurological defects.
Collapse
Affiliation(s)
- Rahul Tripathi
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Mehar Sahu
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Devesh Srivastava
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Ankita Das
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India.
- , Delhi, India.
- Molecular Neuroscience and Functional Genomics Laboratory, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India.
| |
Collapse
|
12
|
Excitatory Synaptic Transmission in Ischemic Stroke: A New Outlet for Classical Neuroprotective Strategies. Int J Mol Sci 2022; 23:ijms23169381. [PMID: 36012647 PMCID: PMC9409263 DOI: 10.3390/ijms23169381] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 01/01/2023] Open
Abstract
Stroke is one of the leading causes of death and disability in the world, of which ischemia accounts for the majority. There is growing evidence of changes in synaptic connections and neural network functions in the brain of stroke patients. Currently, the studies on these neurobiological alterations mainly focus on the principle of glutamate excitotoxicity, and the corresponding neuroprotective strategies are limited to blocking the overactivation of ionic glutamate receptors. Nevertheless, it is disappointing that these treatments often fail because of the unspecificity and serious side effects of the tested drugs in clinical trials. Thus, in the prevention and treatment of stroke, finding and developing new targets of neuroprotective intervention is still the focus and goal of research in this field. In this review, we focus on the whole processes of glutamatergic synaptic transmission and highlight the pathological changes underlying each link to help develop potential therapeutic strategies for ischemic brain damage. These strategies include: (1) controlling the synaptic or extra-synaptic release of glutamate, (2) selectively blocking the action of the glutamate receptor NMDAR subunit, (3) increasing glutamate metabolism, and reuptake in the brain and blood, and (4) regulating the glutamate system by GABA receptors and the microbiota–gut–brain axis. Based on these latest findings, it is expected to promote a substantial understanding of the complex glutamate signal transduction mechanism, thereby providing excellent neuroprotection research direction for human ischemic stroke (IS).
Collapse
|
13
|
Fagerli E, Escobar I, Ferrier FJ, Jackson CW, Perez-Lao EJ, Perez-Pinzon MA. Sirtuins and cognition: implications for learning and memory in neurological disorders. Front Physiol 2022; 13:908689. [PMID: 35936890 PMCID: PMC9355297 DOI: 10.3389/fphys.2022.908689] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/30/2022] [Indexed: 11/18/2022] Open
Abstract
Sirtuins are an evolutionarily conserved family of regulatory proteins that function in an NAD+ -dependent manner. The mammalian family of sirtuins is composed of seven histone deacetylase and ADP-ribosyltransferase proteins (SIRT1-SIRT7) that are found throughout the different cellular compartments of the cell. Sirtuins in the brain have received considerable attention in cognition due to their role in a plethora of metabolic and age-related diseases and their ability to induce neuroprotection. More recently, sirtuins have been shown to play a role in normal physiological cognitive function, and aberrant sirtuin function is seen in pathological cellular states. Sirtuins are believed to play a role in cognition through enhancing synaptic plasticity, influencing epigenetic regulation, and playing key roles in molecular pathways involved with oxidative stress affecting mitochondrial function. This review aims to discuss recent advances in the understanding of the role of mammalian sirtuins in cognitive function and the therapeutic potential of targeting sirtuins to ameliorate cognitive deficits in neurological disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Miguel A. Perez-Pinzon
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
14
|
Philbert SA, Xu J, Church SJ, Unwin RD, Roncaroli F, Cooper GJS. Pan-cerebral sodium elevations in vascular dementia: Evidence for disturbed brain-sodium homeostasis. Front Aging Neurosci 2022; 14:926463. [PMID: 35923550 PMCID: PMC9340791 DOI: 10.3389/fnagi.2022.926463] [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: 04/22/2022] [Accepted: 06/28/2022] [Indexed: 01/31/2023] Open
Abstract
Vascular dementia (VaD) is the second most common cause of cognitive impairment amongst the elderly. However, there are no known disease-modifying therapies for VaD, probably due to incomplete understanding of the molecular basis of the disease. Despite the complex etiology of neurodegenerative conditions, a growing body of research now suggests the potential involvement of metal dyshomeostasis in the pathogenesis of several of the age-related dementias. However, by comparison, there remains little research investigating brain metal levels in VaD. In order to shed light on the possible involvement of metal dyshomeostasis in VaD, we employed inductively coupled plasma-mass spectrometry to quantify the levels of essential metals in post-mortem VaD brain tissue (n = 10) and age-/sex-matched controls (n = 10) from seven brain regions. We found novel evidence for elevated wet-weight cerebral sodium levels in VaD brain tissue in six out of the seven regions analyzed. Decreased cerebral-potassium levels as well as increased Na/K ratios (consistent with high tissue sodium and low potassium levels) were also observed in several brain regions. These data suggest that reduced Na+/K+-exchanging ATPase (EC 7.2.2.13) activity could contribute to the contrasting changes in sodium and potassium measured here.
Collapse
Affiliation(s)
- Sasha A. Philbert
- Division of Cardiovascular Sciences, Centre for Advanced Discovery and Experimental Therapeutics, Faculty of Biology, Medicine and Health, School of Medical Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- *Correspondence: Sasha A. Philbert,
| | - Jingshu Xu
- Division of Cardiovascular Sciences, Centre for Advanced Discovery and Experimental Therapeutics, Faculty of Biology, Medicine and Health, School of Medical Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Stephanie J. Church
- Division of Cardiovascular Sciences, Centre for Advanced Discovery and Experimental Therapeutics, Faculty of Biology, Medicine and Health, School of Medical Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Richard D. Unwin
- Division of Cardiovascular Sciences, Centre for Advanced Discovery and Experimental Therapeutics, Faculty of Biology, Medicine and Health, School of Medical Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Division of Cancer Sciences, Stoller Biomarker Discovery Centre, Faculty of Biology, Medicine and Health, School of Medical Sciences, The University of Manchester, Manchester, United Kingdom
| | - Federico Roncaroli
- Division of Neuroscience and Experimental Psychology, Geoffrey Jefferson Brain Research Centre, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Garth J. S. Cooper
- Division of Cardiovascular Sciences, Centre for Advanced Discovery and Experimental Therapeutics, Faculty of Biology, Medicine and Health, School of Medical Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Faculty of Science, School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
15
|
Rodrigues T, Piccirillo S, Magi S, Preziuso A, Dos Santos Ramos V, Serfilippi T, Orciani M, Maciel Palacio Alvarez M, Luis Dos Santos Tersariol I, Amoroso S, Lariccia V. Control of Ca 2+ and metabolic homeostasis by the Na +/Ca 2+ exchangers (NCXs) in health and disease. Biochem Pharmacol 2022; 203:115163. [PMID: 35803319 DOI: 10.1016/j.bcp.2022.115163] [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: 04/01/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 11/16/2022]
Abstract
Spatial and temporal control of calcium (Ca2+) levels is essential for the background rhythms and responses of living cells to environmental stimuli. Whatever other regulators a given cellular activity may have, localized and wider scale Ca2+ events (sparks, transients, and waves) are hierarchical determinants of fundamental processes such as cell contraction, excitability, growth, metabolism and survival. Different cell types express specific channels, pumps and exchangers to efficiently generate and adapt Ca2+ patterns to cell requirements. The Na+/Ca2+ exchangers (NCXs) in particular contribute to Ca2+ homeostasis by buffering intracellular Ca2+ loads according to the electrochemical gradients of substrate ions - i.e., Ca2+ and sodium (Na+) - and under a dynamic control of redundant regulatory processes. An interesting feature of NCX emerges from the strict relationship that connects transporter activity with cell metabolism: on the one hand NCX operates under constant control of ATP-dependent regulatory processes, on the other hand the ion fluxes generated through NCX provide mechanistic support for the Na+-driven uptake of glutamate and Ca2+ influx to fuel mitochondrial respiration. Proof of concept evidence highlights therapeutic potential of preserving a timed and balanced NCX activity in a growing rate of diseases (including excitability, neurodegenerative, and proliferative disorders) because of an improved ability of stressed cells to safely maintain ion gradients and mitochondrial bioenergetics. Here, we will summarize and review recent works that have focused on the pathophysiological roles of NCXs in balancing the two-way relationship between Ca2+ signals and metabolism.
Collapse
Affiliation(s)
- Tiago Rodrigues
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, SP, Brazil.
| | - Silvia Piccirillo
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Simona Magi
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Alessandra Preziuso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Vyctória Dos Santos Ramos
- Interdisciplinary Center for Biochemistry Investigation (CIIB), University of Mogi das Cruzes (UMC), Mogi das Cruzes, SP, Brazil
| | - Tiziano Serfilippi
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Monia Orciani
- Department of Clinical and Molecular Sciences, Histology, University "Politecnica delle Marche", Ancona, Italy.
| | - Marcela Maciel Palacio Alvarez
- Department of Biochemistry, São Paulo School of Medicine, Federal University of São Paulo (Unifesp) São Paulo, SP, Brazil
| | | | - Salvatore Amoroso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Vincenzo Lariccia
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| |
Collapse
|
16
|
The Evidence for Effective Inhibition of INa Produced by Mirogabalin ((1R,5S,6S)-6-(aminomethyl)-3-ethyl-bicyclo [3.2.0] hept-3-ene-6-acetic acid), a Known Blocker of Ca V Channels. Int J Mol Sci 2022; 23:ijms23073845. [PMID: 35409204 PMCID: PMC8998350 DOI: 10.3390/ijms23073845] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 01/27/2023] Open
Abstract
Mirogabalin (MGB, Tarlige®), an inhibitor of the α2δ-1 subunit of voltage-gated Ca2+ (CaV) channels, is used as a way to alleviate peripheral neuropathic pain and diabetic neuropathy. However, to what extent MGB modifies the magnitude, gating, and/or hysteresis of various types of plasmalemmal ionic currents remains largely unexplored. In pituitary tumor (GH3) cells, we found that MGB was effective at suppressing the peak (transient, INa(T)) and sustained (late, INa(L)) components of the voltage-gated Na+ current (INa) in a concentration-dependent manner, with an effective IC50 of 19.5 and 7.3 μM, respectively, while the KD value calculated on the basis of minimum reaction scheme was 8.2 μM. The recovery of INa(T) inactivation slowed in the presence of MGB, although the overall current-voltage relation of INa(T) was unaltered; however, there was a leftward shift in the inactivation curve of the current. The magnitude of the window (INa(W)) or resurgent INa (INa(R)) evoked by the respective ascending or descending ramp pulse (Vramp) was reduced during cell exposure to MGB. MGB-induced attenuation in INa(W) or INa(R) was reversed by the further addition of tefluthrin, a pyrethroid insecticide known to stimulate INa. MGB also effectively lessened the strength of voltage-dependent hysteresis of persistent INa in response to the isosceles triangular Vramp. The cumulative inhibition of INa(T), evoked by pulse train stimulation, was enhanced in its presence. Taken together, in addition to the inhibition of CaV channels, the NaV channel attenuation produced by MGB might have an impact in its analgesic effects occurring in vivo.
Collapse
|
17
|
Pluta R, Kiś J, Januszewski S, Jabłoński M, Czuczwar SJ. Cross-Talk between Amyloid, Tau Protein and Free Radicals in Post-Ischemic Brain Neurodegeneration in the Form of Alzheimer’s Disease Proteinopathy. Antioxidants (Basel) 2022; 11:antiox11010146. [PMID: 35052650 PMCID: PMC8772936 DOI: 10.3390/antiox11010146] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/05/2022] [Accepted: 01/09/2022] [Indexed: 02/04/2023] Open
Abstract
Recent years have seen remarkable progress in research into free radicals oxidative stress, particularly in the context of post-ischemic recirculation brain injury. Oxidative stress in post-ischemic tissues violates the integrity of the genome, causing DNA damage, death of neuronal, glial and vascular cells, and impaired neurological outcome after brain ischemia. Indeed, it is now known that DNA damage and repair play a key role in post-stroke white and gray matter remodeling, and restoring the integrity of the blood-brain barrier. This review will present one of the newly characterized mechanisms that emerged with genomic and proteomic development that led to brain ischemia to a new level of post-ischemic neuropathological mechanisms, such as the presence of amyloid plaques and the development of neurofibrillary tangles, which further exacerbate oxidative stress. Finally, we hypothesize that modified amyloid and the tau protein, along with the oxidative stress generated, are new key elements in the vicious circle important in the development of post-ischemic neurodegeneration in a type of Alzheimer’s disease proteinopathy.
Collapse
Affiliation(s)
- Ryszard Pluta
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 Str., 02-106 Warsaw, Poland;
- Correspondence: ; Tel.: +48-22-608-6540
| | - Jacek Kiś
- Department of Urology, 1st Military Clinical Hospital with the Outpatient Clinic, Al. Racławickie 23, 20-049 Lublin, Poland;
| | - Sławomir Januszewski
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 Str., 02-106 Warsaw, Poland;
| | - Mirosław Jabłoński
- Department of Rehabilitation and Orthopedics, Medical University of Lublin, Jaczewskiego 8 Str., 20-090 Lublin, Poland;
| | - Stanisław J. Czuczwar
- Department of Pathophysiology, Medical University of Lublin, Jaczewskiego 8b Str., 20-090 Lublin, Poland;
| |
Collapse
|
18
|
Yu Q, Liu W, Chen Z, Zhang M. Specificity Protein 1: A Protein With a Two-Sided Role in Ischemic Stroke. Front Cell Neurosci 2022; 15:757670. [PMID: 34970121 PMCID: PMC8712767 DOI: 10.3389/fncel.2021.757670] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/15/2021] [Indexed: 11/23/2022] Open
Abstract
Stroke is one of the leading causes of death and disability worldwide. However, there is a lack of effective medications to speed up the recovery process. Ischemic stroke, as the result of cerebral infarction or cerebral artery narrowing, is accompanied by hemiplegia or impaired consciousness. There are many transcription factors involved in the development of this condition, whose alterations can influence or signal the prognostic outcomes of ischemic stroke. Among them, the augmented expression of specificity protein 1 (SP1) can participate in the progression of the disease by binding DNA to regulate the transcriptions of many genes. Different studies have provided different answers as to whether SP1 plays a positive or a negative role in ischemic stroke. On the one hand, SP1 can play a cytoprotective role as both an antioxidant and anti-apoptotic agent for neurons and glial cells. On the other hand, it can also damage neuronal cells by promoting inflammation and exacerbating brain edema. In this review, we highlight the roles of SP1 in ischemic stroke and shed light on the underlying mechanism.
Collapse
Affiliation(s)
- Qinyang Yu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wangyang Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhuohui Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Mengqi Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
19
|
Costas-Ferreira C, Faro LRF. Systematic Review of Calcium Channels and Intracellular Calcium Signaling: Relevance to Pesticide Neurotoxicity. Int J Mol Sci 2021; 22:13376. [PMID: 34948173 PMCID: PMC8704302 DOI: 10.3390/ijms222413376] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/25/2022] Open
Abstract
Pesticides of different chemical classes exert their toxic effects on the nervous system by acting on the different regulatory mechanisms of calcium (Ca2+) homeostasis. Pesticides have been shown to alter Ca2+ homeostasis, mainly by increasing its intracellular concentration above physiological levels. The pesticide-induced Ca2+ overload occurs through two main mechanisms: the entry of Ca2+ from the extracellular medium through the different types of Ca2+ channels present in the plasma membrane or its release into the cytoplasm from intracellular stocks, mainly from the endoplasmic reticulum. It has also been observed that intracellular increases in the Ca2+ concentrations are maintained over time, because pesticides inhibit the enzymes involved in reducing its levels. Thus, the alteration of Ca2+ levels can lead to the activation of various signaling pathways that generate oxidative stress, neuroinflammation and, finally, neuronal death. In this review, we also discuss some proposed strategies to counteract the detrimental effects of pesticides on Ca2+ homeostasis.
Collapse
Affiliation(s)
| | - Lilian R. F. Faro
- Departamento de Biología Funcional y Ciencias de la Salud, Facultad de Biología, Universidade de Vigo, Campus Universitario As Lagoas Marcosende, 36310 Vigo, Spain;
| |
Collapse
|
20
|
Liu Y, Cheng Y, Zhang W, Tian H. Neuroprotective Effect of a New Free Radical Scavenger HL-008 in an Ischemia-Reperfusion Injury Rat Model. Neuroscience 2021; 465:105-115. [PMID: 33915200 DOI: 10.1016/j.neuroscience.2021.04.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 12/28/2022]
Abstract
Oxidative stress plays a critical role in cerebral ischemia-reperfusion injury. We have previously developed a powerful antioxidant, HL-008. This study aimed to investigate the neuroprotective function of HL-008. HL-008 efficacy in vitro and in vivo was evaluated using a PC-12 cell oxidative stress model induced by hydrogen peroxide and a rat model of middle cerebral artery occlusion, respectively. The MTT assay was used to analyze cell viability. 2,3,5-Triphenyltetrazolium chloride and Hematoxylin and Eosin staining, immunofluorescence, western blot, and proteomics were used to evaluate the infarction volume, brain tissue morphology, apoptosis, inflammation, and related pathways. Indicators related to oxidative levels were also detected. HL-008 significantly reduced the cerebral infarction volume induced by ischemia-reperfusion, improved the neurological score, alleviated oxidative stress and inflammation in the brain tissue, reduced glial cell activation, inhibited brain tissue apoptosis by influencing multiple signaling pathways, and had a neuroprotective effect. If HL-008 is successfully developed, it could significantly improve stroke patients' quality of life.
Collapse
Affiliation(s)
- Yahong Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 238, Baidi Road, Tianjin, China
| | - Ying Cheng
- Center for Marine Bioproducts Development, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, SA 5042, Australia
| | - Wei Zhang
- Center for Marine Bioproducts Development, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, SA 5042, Australia.
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 238, Baidi Road, Tianjin, China.
| |
Collapse
|
21
|
Shi ZF, Fang Q, Chen Y, Xu LX, Wu M, Jia M, Lu Y, Wang XX, Wang YJ, Yan X, Dong LP, Yuan F. Methylene blue ameliorates brain edema in rats with experimental ischemic stroke via inhibiting aquaporin 4 expression. Acta Pharmacol Sin 2021; 42:382-392. [PMID: 32665706 PMCID: PMC8027449 DOI: 10.1038/s41401-020-0468-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/23/2020] [Indexed: 12/23/2022] Open
Abstract
Brain edema is a common and serious complication of ischemic stroke with limited effective treatment. We previously reported that methylene blue (MB) attenuated ischemic brain edema in rats, but the underlying mechanisms remained unknown. Aquaporin 4 (AQP4) in astrocytes plays a key role in brain edema. We also found that extracellular signal-regulated kinase 1/2 (ERK1/2) activation was involved in the regulation of AQP4 expression in astrocytes. In the present study, we investigated whether AQP4 and ERK1/2 were involved in the protective effect of MB against cerebral edema. Rats were subjected to transient middle cerebral artery occlusion (tMCAO), MB (3 mg/kg, for 30 min) was infused intravenously through the tail vein started immediately after reperfusion and again at 3 h after ischemia (1.5 mg/kg, for 15 min). Brain edema was determined by MRI at 0.5, 2.5, and 48 h after tMCAO. The decreases of apparent diffusion coefficient (ADC) values on diffusion-weighted MRI indicated cytotoxic brain edema, whereas the increase of T2 MRI values reflected vasogenic brain edema. We found that MB infusion significantly ameliorated cytotoxic brain edema at 2.5 and 48 h after tMCAO and decreased vasogenic brain edema at 48 h after tMCAO. In addition, MB infusion blocked the AQP4 increases and ERK1/2 activation in the cerebral cortex in ischemic penumbra at 48 h after tMCAO. In a cell swelling model established in cultured rat astrocyte exposed to glutamate (1 mM), we consistently found that MB (10 μM) attenuated cell swelling, AQP4 increases and ERK1/2 activation. Moreover, the ERK1/2 inhibitor U0126 (10 μM) had the similar effects as MB. These results demonstrate that MB improves brain edema and astrocyte swelling, which may be mediated by the inhibition of AQP4 expression via ERK1/2 pathway, suggesting that MB may be a potential choice for the treatment of brain edema.
Collapse
Affiliation(s)
- Zhong-Fang Shi
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- Beijing Key Laboratory of Central Nervous System Injury, Beijing, 100070, China
| | - Qing Fang
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Ye Chen
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Li-Xin Xu
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Min Wu
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Mei Jia
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Yi Lu
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Xiao-Xuan Wang
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Yu-Jiao Wang
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Xu Yan
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Li-Ping Dong
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Fang Yuan
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
- Beijing Key Laboratory of Central Nervous System Injury, Beijing, 100070, China.
| |
Collapse
|
22
|
Chiarantin GMD, Delgado-Garcia LM, Zamproni LN, Lima MA, Nader HB, Tersariol ILS, Porcionatto M. Neuroprotective effect of heparin Trisulfated disaccharide on ischemic stroke. Glycoconj J 2021; 38:35-43. [PMID: 33411076 DOI: 10.1007/s10719-020-09966-4] [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: 08/05/2020] [Revised: 11/15/2020] [Accepted: 11/26/2020] [Indexed: 10/22/2022]
Abstract
Cells undergoing hypoxia experience intense cytoplasmic calcium (Ca2+) overload. High concentrations of intracellular calcium ([Ca2+]i) can trigger cell death in the neural tissue, a hallmark of stroke. Neural Ca2+ homeostasis involves regulation by the Na+/Ca2+ exchanger (NCX). Previous data published by our group showed that a product of the enzymatic depolymerization of heparin by heparinase, the unsaturated trisulfated disaccharide (TD; ΔU, 2S-GlcNS, 6S), can accelerate Na+/Ca2+ exchange via NCX, in hepatocytes and aorta vascular smooth muscle cells. Thus, the objective of this work was to verify whether TD could act as a neuroprotective agent able to prevent neuronal cell death by reducing [Ca2+]i. Pretreatment of N2a cells with TD reduced [Ca2+]i rise induced by thapsigargin and increased cell viability under [Ca2+]I overload conditions and in hypoxia. Using a murine model of stroke, we observed that pretreatment with TD decreased cerebral infarct volume and cell death. However, when mice received KB-R7943, an NCX blocker, the neuroprotective effect of TD was abolished, strongly suggesting that this neuroprotection requires a functional NCX to happen. Thus, we propose TD-NCX as a new therapeutic axis for the prevention of neuronal death induced by [Ca2+]i overload.
Collapse
Affiliation(s)
- Gabrielly M D Chiarantin
- Laboratory of Molecular Neurobiology, Universidade Federal de São Paulo, São Paulo, SP, Brazil
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Lina M Delgado-Garcia
- Laboratory of Molecular Neurobiology, Universidade Federal de São Paulo, São Paulo, SP, Brazil
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Laura N Zamproni
- Laboratory of Molecular Neurobiology, Universidade Federal de São Paulo, São Paulo, SP, Brazil
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Marcelo A Lima
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, SP, Brazil
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Newcastle-Under-Lyme, Staffordshire, ST5 5BG, UK
| | - Helena B Nader
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Ivarne L S Tersariol
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| | - Marimélia Porcionatto
- Laboratory of Molecular Neurobiology, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
23
|
Drug development in targeting ion channels for brain edema. Acta Pharmacol Sin 2020; 41:1272-1288. [PMID: 32855530 PMCID: PMC7609292 DOI: 10.1038/s41401-020-00503-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/02/2020] [Indexed: 12/18/2022] Open
Abstract
Cerebral edema is a pathological hallmark of various central nervous system (CNS) insults, including traumatic brain injury (TBI) and excitotoxic injury such as stroke. Due to the rigidity of the skull, edema-induced increase of intracranial fluid significantly complicates severe CNS injuries by raising intracranial pressure and compromising perfusion. Mortality due to cerebral edema is high. With mortality rates up to 80% in severe cases of stroke, it is the leading cause of death within the first week. Similarly, cerebral edema is devastating for patients of TBI, accounting for up to 50% mortality. Currently, the available treatments for cerebral edema include hypothermia, osmotherapy, and surgery. However, these treatments only address the symptoms and often elicit adverse side effects, potentially in part due to non-specificity. There is an urgent need to identify effective pharmacological treatments for cerebral edema. Currently, ion channels represent the third-largest target class for drug development, but their roles in cerebral edema remain ill-defined. The present review aims to provide an overview of the proposed roles of ion channels and transporters (including aquaporins, SUR1-TRPM4, chloride channels, glucose transporters, and proton-sensitive channels) in mediating cerebral edema in acute ischemic stroke and TBI. We also focus on the pharmacological inhibitors for each target and potential therapeutic strategies that may be further pursued for the treatment of cerebral edema.
Collapse
|
24
|
Zhao Y, Yang J, Li C, Zhou G, Wan H, Ding Z, Wan H, Zhou H. Role of the neurovascular unit in the process of cerebral ischemic injury. Pharmacol Res 2020; 160:105103. [PMID: 32739425 DOI: 10.1016/j.phrs.2020.105103] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/17/2022]
Abstract
Cerebral ischemic injury exhibits both high morbidity and mortality worldwide. Traditional research of the pathogenesis of cerebral ischemic injury has focused on separate analyses of the involved cell types. In recent years, the neurovascular unit (NVU) mechanism of cerebral ischemic injury has been proposed in modern medicine. Hence, more effective strategies for the treatment of cerebral ischemic injury may be provided through comprehensive analysis of brain cells and the extracellular matrix. However, recent studies that have investigated the function of the NVU in cerebral ischemic injury have been insufficient. In addition, the metabolism and energy conversion of the NVU depend on interactions among multiple cell types, which make it difficult to identify the unique contribution of each cell type. Therefore, in the present review, we comprehensively summarize the regulatory effects and recovery mechanisms of four major cell types (i.e., astrocytes, microglia, brain-microvascular endothelial cells, and neurons) in the NVU under cerebral ischemic injury, as well as discuss the interactions among these cell types in the NVU. Furthermore, we discuss the common signaling pathways and signaling factors that mediate cerebral ischemic injury in the NVU, which may help to provide a theoretical basis for the comprehensive elucidation of cerebral ischemic injury.
Collapse
Affiliation(s)
- Yu Zhao
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Jiehong Yang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Chang Li
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Guoying Zhou
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Haofang Wan
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Zhishan Ding
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Haitong Wan
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| | - Huifen Zhou
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| |
Collapse
|
25
|
Piccirillo S, Magi S, Castaldo P, Preziuso A, Lariccia V, Amoroso S. NCX and EAAT transporters in ischemia: At the crossroad between glutamate metabolism and cell survival. Cell Calcium 2020; 86:102160. [PMID: 31962228 DOI: 10.1016/j.ceca.2020.102160] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/10/2020] [Accepted: 01/10/2020] [Indexed: 01/29/2023]
Abstract
Energy metabolism impairment is a central event in the pathophysiology of ischemia. The limited availability of glucose and oxygen strongly affects mitochondrial activity, thus leading to ATP depletion. In this setting, the switch to alternative energy sources could ameliorate cells survival by enhancing ATP production, thus representing an attractive strategy for ischemic treatment. In this regard, some studies have recently re-evaluated the metabolic role of glutamate and its potential to promote cell survival under pathological conditions. In the present review, we discuss the ability of glutamate to exert an "energizing role" in cardiac and neuronal models of hypoxia/reoxygenation (H/R) injury, focusing on the Na+/Ca2+ exchanger (NCX) and the Na+-dependent excitatory amino acid transporters (EAATs) as key players in this metabolic pathway.
Collapse
Affiliation(s)
- Silvia Piccirillo
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126, Ancona, Italy
| | - Simona Magi
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126, Ancona, Italy.
| | - Pasqualina Castaldo
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126, Ancona, Italy
| | - Alessandra Preziuso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126, Ancona, Italy
| | - Vincenzo Lariccia
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126, Ancona, Italy
| | - Salvatore Amoroso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126, Ancona, Italy
| |
Collapse
|
26
|
Anuriev AM, Gorbachev VI. [Hypoxic-ischemic brain damage in premature newborns]. Zh Nevrol Psikhiatr Im S S Korsakova 2020; 119:63-69. [PMID: 31825364 DOI: 10.17116/jnevro201911908263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
One of the main causes of cerebral dysfunction in premature newborns is hypoxia. High mortality and lifelong morbidity in these children is a frequent result of neonatal hypoxic brain damage. The article presents some data on the prevalence of neurological diseases that have arisen in the perinatal period, and highlights the key etiological factors leading to hypoxia in both the intranatal and early postnatal periods. The pathogenesis of hypoxic-ischemic brain lesions in premature infants is described in detail. At the same time, more careful consideration is given to the glutathione system, which protects against lipid peroxidation, the glutamate-calcium cascade, and the excitotoxicity mediated by it, as well as the processes of necrosis and apoptosis of nerve cells. The advantages and disadvantages of modern methods for diagnosing cerebral lesions are noted, and the principles of treatment of these disorders are analyzed.
Collapse
Affiliation(s)
- A M Anuriev
- Irkutsk State Medical Academy of Postgraduate Education - Branch Campus of the Russian Medical Academy of Continuing Professional Education, Irkutsk, Russia
| | - V I Gorbachev
- Irkutsk State Medical Academy of Postgraduate Education - Branch Campus of the Russian Medical Academy of Continuing Professional Education, Irkutsk, Russia
| |
Collapse
|
27
|
Song S, Luo L, Sun B, Sun D. Roles of glial ion transporters in brain diseases. Glia 2019; 68:472-494. [PMID: 31418931 DOI: 10.1002/glia.23699] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/22/2019] [Accepted: 07/26/2019] [Indexed: 12/21/2022]
Abstract
Glial ion transporters are important in regulation of ionic homeostasis, cell volume, and cellular signal transduction under physiological conditions of the central nervous system (CNS). In response to acute or chronic brain injuries, these ion transporters can be activated and differentially regulate glial functions, which has subsequent impact on brain injury or tissue repair and functional recovery. In this review, we summarized the current knowledge about major glial ion transporters, including Na+ /H+ exchangers (NHE), Na+ /Ca2+ exchangers (NCX), Na+ -K+ -Cl- cotransporters (NKCC), and Na+ -HCO3 - cotransporters (NBC). In acute neurological diseases, such as ischemic stroke and traumatic brain injury (TBI), these ion transporters are rapidly activated and play significant roles in regulation of the intra- and extracellular pH, Na+ , K+ , and Ca2+ homeostasis, synaptic plasticity, and myelin formation. However, overstimulation of these ion transporters can contribute to glial apoptosis, demyelination, inflammation, and excitotoxicity. In chronic brain diseases, such as glioma, Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS), glial ion transporters are involved in the glioma Warburg effect, glial activation, neuroinflammation, and neuronal damages. These findings suggest that glial ion transporters are involved in tissue structural and functional restoration, or brain injury and neurological disease development and progression. A better understanding of these ion transporters in acute and chronic neurological diseases will provide insights for their potential as therapeutic targets.
Collapse
Affiliation(s)
- Shanshan Song
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lanxin Luo
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, Pennsylvania.,School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China.,School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Baoshan Sun
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China.,Pólo Dois Portos, Instituto National de Investigação Agrária e Veterinária, Dois Portos, Portugal
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, Pennsylvania.,Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational and Clinical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
28
|
Hu HJ, Wang SS, Wang YX, Liu Y, Feng XM, Shen Y, Zhu L, Chen HZ, Song M. Blockade of the forward Na + /Ca 2+ exchanger suppresses the growth of glioblastoma cells through Ca 2+ -mediated cell death. Br J Pharmacol 2019; 176:2691-2707. [PMID: 31034096 PMCID: PMC6609550 DOI: 10.1111/bph.14692] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 01/13/2023] Open
Abstract
Background and Purpose The Na+/Ca2+ exchanger (NCX) working in either forward or reverse mode participates in maintaining intracellular Ca2+ ([Ca2+]i) homeostasis, which is essential for determining cell fate. Previously, numerous blockers targeting reverse or forward NCX have been developed and studied in ischaemic tissue injury but barely examined in glioblastoma for the purpose of anti‐tumour therapy. We assessed the effect of NCX blockers on glioblastoma growth and whether NCX can become a therapeutic target. Experimental Approach Patch‐clamp recording, Ca2+ imaging, flow cytometry, and Western blot were used to study the effects of specific and non‐specific NCX blockers on cultured glioblastoma cells. In vivo bioluminescent imaging was used to measure effects on grafted glioblastoma. Key Results Selectively blocking the reverse NCX with SEA0400, SN‐6, and YM‐244769 did not affect tumour cell viability. Blocking the forward NCX with bepridil, CB‐DMB, or KB‐R7943 elevated [Ca2+]i and killed glioblastoma cells. Bepridil and CB‐DMB caused Ca2+‐dependent cell cycle arrest together with apoptosis, which were all attenuated by a Ca2+ chelator BAPTA‐AM. Systemic administration of bepridil inhibited growth of brain‐grafted glioblastoma. Bepridil did not appear to have a cytotoxic effect on human astrocytes, which have higher functional expression of NCX than glioblastoma cells. Conclusions and Implications Low expression of the NCX makes glioblastoma cells sensitive to disturbance of [Ca2+]i. Interventions designed to block the forward NCX can cause Ca2+‐mediated injury to glioblastoma thus having therapeutic potential. Bepridil could be a lead compound for developing new anti‐tumour drugs.
Collapse
Affiliation(s)
- Hui-Jie Hu
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shan-Shan Wang
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan-Xia Wang
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Liu
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue-Mei Feng
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Shen
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Zhu
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong-Zhuan Chen
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingke Song
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Moriguchi S, Kita S, Iwamoto T, Fukunaga K. [Dysfunction of Na +/Ca 2+ exchangers is associated with cognitive decline in Alzheimer's disease]. Nihon Yakurigaku Zasshi 2018; 152:299-305. [PMID: 30531101 DOI: 10.1254/fpj.152.299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Na+/Ca2+ exchanger (NCX) is mainly expressed in the plasma membrane and mediates electrogenical exchange of one Ca2+ for three Na+, depending on the electrochemical gradients across the plasma membrane. NCX has three different isoforms (NCX1, NCX2, NCX3) encoded by distinct genes in mammals. Here, we report that NCX2 and NCX3 protein levels are relatively reduced in hippocampal CA1 of Alzheimer's disease model mice. Likewise, NCX2+/- or NCX3+/- mice exhibited impaired hippocampal LTP and memory-related behaviors. In immunoblot analyses, calcium/calmodulin-dependent protein kinase II (CaMKII) autophosphorylation significantly decreased in hippocampal CA1 of NCX2+/- mice compared to wild-type mice. By contrast, NCX2+/- mice was correlated with elevated calcineurin (CaN) activity and rescued by treatment with the calcineurin inhibitor FK506. Taken together, the imbalance of CaMKII and CaN activities with concomitant LTP impairment likely accounts for the learning disability observed in NCX2+/- mice.
Collapse
Affiliation(s)
- Shigeki Moriguchi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University
| | - Satomi Kita
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University.,Department of Pharmacology, Faculty of Medicine, Fukuoka University
| | - Takahiro Iwamoto
- Department of Pharmacology, Faculty of Medicine, Fukuoka University
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University
| |
Collapse
|
30
|
Ostrowski RP, Zhang JH. The insights into molecular pathways of hypoxia-inducible factor in the brain. J Neurosci Res 2018; 98:57-76. [PMID: 30548473 DOI: 10.1002/jnr.24366] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 11/16/2018] [Accepted: 11/20/2018] [Indexed: 12/12/2022]
Abstract
The objectives of this present work were to review recent developments on the role of hypoxia-inducible factor (HIF) in the survival of cells under normoxic versus hypoxic and inflammatory brain conditions. The dual nature of HIF effects appears well established, based on the accumulated evidence of HIF playing both the role of adaptive factor and mediator of cell demise. Cellular HIF responses depend on pathophysiological conditions, developmental phase, comorbidities, and administered medications. In addition, HIF-1α and HIF-2α actions may vary in the same tissues. The multiple roles of HIF in stem cells are emerging. HIF not only regulates expression of target genes and thereby influences resultant protein levels but also contributes to epigenetic changes that may reciprocally provide feedback regulations loops. These HIF-dependent alterations in neurological diseases and its responses to treatments in vivo need to be examined alongside with a functional status of subjects involved in such studies. The knowledge of HIF pathways might be helpful in devising HIF-mimetics and modulating drugs, acting on the molecular level to improve clinical outcomes, as exemplified here by clinical and experimental data of selected brain diseases, occasionally corroborated by the data from disorders of other organs. Because of complex role of HIF in brain injuries, prospective therapeutic interventions need to differentially target HIF responses depending on their roles in the molecular mechanisms of neurologic diseases.
Collapse
Affiliation(s)
- Robert P Ostrowski
- Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - John H Zhang
- Departments of Anesthesiology and Physiology, School of Medicine, Loma Linda University, Loma Linda, California
| |
Collapse
|
31
|
Moriguchi S, Kita S, Inagaki R, Yabuki Y, Sasaki Y, Ishikawa S, Sakagami H, Iwamoto T, Fukunaga K. Aberrant Amygdala-Dependent Cued Fear Memory in Na +/Ca 2+ Exchanger 1 Heterozygous Mice. Mol Neurobiol 2018; 56:4381-4394. [PMID: 30324228 DOI: 10.1007/s12035-018-1384-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 10/08/2018] [Indexed: 02/06/2023]
Abstract
Na+/Ca2+ exchangers (NCXs) are mainly expressed in the plasma membrane and exchange one Ca2+ for three Na+, depending on the electrochemical gradients across the plasma membrane. NCXs have three isoforms, NCX1-3, encoded by distinct genes in mammals. Here, we report that heterozygous mice lacking NCX1 (NCX1+/-) exhibit impaired amygdala-dependent cued fear memory. NCX1+/- mice showed significant impairment in fear-related behaviors measured with the elevated-plus maze, light-dark, open-field, and marble-burying tasks. In addition, NCX1+/- mice showed abnormality in cued fear memory but not in contextual fear memory in a fear-conditioning task. In immunohistochemical analyses, NCX1+/- mice had significantly increased number of c-Fos-positive cells in the lateral amygdala (LA) but not in the central amygdala following fear-related tone stimuli. c-Fos expression peaked at 1 h. In concordance with the aberrant fear-related behaviors in NCX1+/- mice, enhanced long-term potentiation was also observed in the LA of these mice. Furthermore, enhancement of CaMKII or CaMKIV activity in the LA was observed in NCX1+/- mice by immunoblot analyses. In contrast, CaMKII+/- but not CaMKIV-/- mice insufficiently exhibited tone-induced cued fear memory and there was no increase in the number of c-Fos-positive cells in the LA. Altogether, the increased CaMKII activity and consequent c-Fos expression likely account for the dysregulation of amygdala-dependent cued fear memory in NCX1+/- mice.
Collapse
Affiliation(s)
- Shigeki Moriguchi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi, 980-8578, Japan.
| | - Satomi Kita
- Department of Pharmacology, Faculty of Medicine, Fukuoka University, 7-45-1, Nanakuma, Jonan-ku, Fukuoka, Fukuoka, 814-0180, Japan.,Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan
| | - Ryo Inagaki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Yasushi Yabuki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Yuzuru Sasaki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Shun Ishikawa
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Hiroyuki Sakagami
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Japan
| | - Takahiro Iwamoto
- Department of Pharmacology, Faculty of Medicine, Fukuoka University, 7-45-1, Nanakuma, Jonan-ku, Fukuoka, Fukuoka, 814-0180, Japan.
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| |
Collapse
|
32
|
Glutamate as a potential "survival factor" in an in vitro model of neuronal hypoxia/reoxygenation injury: leading role of the Na +/Ca 2+ exchanger. Cell Death Dis 2018; 9:731. [PMID: 29955038 PMCID: PMC6023866 DOI: 10.1038/s41419-018-0784-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/04/2018] [Accepted: 06/11/2018] [Indexed: 12/23/2022]
Abstract
In brain ischemia, reduction in oxygen and substrates affects mitochondrial respiratory chain and aerobic metabolism, culminating in ATP production impairment, ionic imbalance, and cell death. The restoration of blood flow and reoxygenation are frequently associated with exacerbation of tissue injury, giving rise to ischemia/reperfusion (I/R) injury. In this setting, the imbalance of brain bioenergetics induces important metabolic adaptations, including utilization of alternative energy sources, such as glutamate. Although glutamate has long been considered as a neurotoxin, it can also be used as intermediary metabolite for ATP synthesis, and both the Na+/Ca2+ exchanger (NCX) and the Na+-dependent excitatory amino-acid transporters (EAATs) are essential in this pathway. Here we analyzed the role of NCX in the potential of glutamate to improve metabolism and survival of neuronal cells subjected to hypoxia/reoxygenation (H/R). In SH-SY5Y neuroblastoma cells differentiated into a neuron-like state, H/R produced a significant cell damage, a decrease in ATP cellular content, and intracellular Ca2+ alterations. Exposure to glutamate at the onset of the reoxygenation phase attenuated H/R-induced cell damage and evoked a significant raise in intracellular ATP levels. Furthermore, we found that in H/R cells NCX reverse-mode activity was reduced, and that glutamate limited such reduction. All the effects induced by glutamate supplementation were lost when cells were transfected with small interfering RNA against NCX1 and EAAT3, suggesting the need of a specific functional interplay between these proteins for glutamate-induced protection. Collectively, our results revealed the potential beneficial effect of glutamate in an in vitro model of H/R injury and focused on the essential role exerted by NCX1. Although preliminary, these findings could be a starting point to further investigate in in vivo systems such protective effect in ischemic settings, shedding a new light on the classical view of glutamate as detrimental factor.
Collapse
|
33
|
Free Radical Damage in Ischemia-Reperfusion Injury: An Obstacle in Acute Ischemic Stroke after Revascularization Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3804979. [PMID: 29770166 PMCID: PMC5892600 DOI: 10.1155/2018/3804979] [Citation(s) in RCA: 307] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/07/2017] [Indexed: 12/16/2022]
Abstract
Acute ischemic stroke is a common cause of morbidity and mortality worldwide. Thrombolysis with recombinant tissue plasminogen activator and endovascular thrombectomy are the main revascularization therapies for acute ischemic stroke. However, ischemia-reperfusion injury after revascularization therapy can result in worsening outcomes. Among all possible pathological mechanisms of ischemia-reperfusion injury, free radical damage (mainly oxidative/nitrosative stress injury) has been found to play a key role in the process. Free radicals lead to protein dysfunction, DNA damage, and lipid peroxidation, resulting in cell death. Additionally, free radical damage has a strong connection with inducing hemorrhagic transformation and cerebral edema, which are the major complications of revascularization therapy, and mainly influencing neurological outcomes due to the disruption of the blood-brain barrier. In order to get a better clinical prognosis, more and more studies focus on the pharmaceutical and nonpharmaceutical neuroprotective therapies against free radical damage. This review discusses the pathological mechanisms of free radicals in ischemia-reperfusion injury and adjunctive neuroprotective therapies combined with revascularization therapy against free radical damage.
Collapse
|
34
|
Ni H, Rui Q, Li D, Gao R, Chen G. The Role of IRE1 Signaling in the Central Nervous System Diseases. Curr Neuropharmacol 2018; 16:1340-1347. [PMID: 29663887 PMCID: PMC6251047 DOI: 10.2174/1570159x16666180416094646] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 08/07/2017] [Accepted: 02/28/2018] [Indexed: 01/14/2023] Open
Abstract
The accumulation of misfolded or unfolded proteins in endoplasmic reticulum (ER) lumen results in the activation of an adaptive stress process called the unfolded protein response (UPR). As the most conserved signaling branch of the UPR, Inositol-requiring enzyme 1 (IRE1) possesses both Ser/Thr kinase and RNase activities operating as major stress sensors, mediating both adaptive and pro-apoptotic pathways under ER stress. Over the last three decades, a mounting body of evidence has shown that IRE1 signaling dysfunction is involved in the pathology of various neurological disorders. Targeting this pathway has emerged as a promising therapeutic strategy against these diseases. In this review, we provide a general overview about the expression and physiological function of IRE1 signaling and its pathophysiological roles in the central nervous system diseases.
Collapse
Affiliation(s)
| | | | - Di Li
- Address correspondence to this author at the Department of Translational Medicine Center, The First People’s Hospital of Zhangjiagang City, Suzhou, Jiangsu, P.R. China; Tel: +86-18921962599; E-mail:
| | | | | |
Collapse
|
35
|
Reduced expression of Na +/Ca 2+ exchangers is associated with cognitive deficits seen in Alzheimer's disease model mice. Neuropharmacology 2017; 131:291-303. [PMID: 29274751 DOI: 10.1016/j.neuropharm.2017.12.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 12/08/2017] [Accepted: 12/20/2017] [Indexed: 11/21/2022]
Abstract
Na+/Ca2+ exchangers (NCXs) are expressed primarily in the plasma membrane of most cell types, where they mediate electrogenic exchange of one Ca2+ for three Na+ ions, depending on Ca2+ and Na+ electrochemical gradients across the membrane. Three mammalian NCX isoforms (NCX1, NCX2, and NCX3) are each encoded by a distinct gene. Here, we report that NCX2 and NCX3 protein and mRNA levels are relatively reduced in hippocampal CA1 of APP23 and APP-KI mice. Likewise, NCX2+/- or NCX3+/- mice exhibited impaired hippocampal LTP and memory-related behaviors. Moreover, relative to controls, calcium/calmodulin-dependent protein kinase II (CaMKII) autophosphorylation significantly decreased in NCX2+/- mouse hippocampus but increased in hippocampus of NCX3+/- mice. NCX2 or NCX3 heterozygotes displayed impaired maintenance of hippocampal LTP, a phenotype that in NCX2+/- mice was correlated with elevated calcineurin activity and rescued by treatment with the calcineurin (CaN) inhibitor FK506. Likewise, FK506 treatment significantly restored impaired hippocampal LTP in APP-KI mice. Moreover, Ca2+ clearance after depolarization following high frequency stimulation was slightly delayed in hippocampal CA1 regions of NCX2+/- mice. Electron microscopy revealed relatively decreased synaptic density in CA1 of NCX2+/- mice, while the number of spines with perforated synapses in CA1 significantly increased in NCX3+/- mice. We conclude that memory impairment seen in NCX2+/- and NCX3+/- mice reflect dysregulated hippocampal CaMKII activity, which alters dendritic spine morphology, findings with implications for memory deficits seen in Alzheimer's disease model mice.
Collapse
|
36
|
Yue JK, Rick JW, Deng H, Feldman MJ, Winkler EA. Efficacy of decompressive craniectomy in the management of intracranial pressure in severe traumatic brain injury. J Neurosurg Sci 2017; 63:425-440. [PMID: 29115100 DOI: 10.23736/s0390-5616.17.04133-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Traumatic brain injury (TBI) is a common cause of permanent disability for which clinical management remains suboptimal. Elevated intracranial pressure (ICP) is a common sequela following TBI leading to death and permanent disability if not properly managed. While clinicians often employ stepwise acute care algorithms to reduce ICP, a number of patients will fail medical management and may be considered for surgical decompression. Decompressive craniectomy (DC) involves removing a component of the bony skull to allow cerebral tissue expansion in order to reduce ICP. However, the impact of DC, which is performed in the setting of neurological instability, ongoing secondary injury, and patient resuscitation, has been challenging to study and outcomes are not well understood. This review summarizes historical and recent studies to elucidate indications for DC and the nuances, risks and complications in its application. The pathophysiology driving ICP elevation, and the corresponding medical interventions for their temporization and treatment, are thoroughly described. The current state of DC - including appropriate injury classification, surgical techniques, concurrent medical therapies, mortality and functional outcomes - is presented. We also report on the recent updates from large randomized controlled trials in severe TBI (Decompressive Craniectomy [DECRA] and Randomized Evaluation of Surgery with Craniectomy for Uncontrollable Elevation of ICP [RESCUEicp]), and recommendations for early DC to treat refractory ICP elevations in malignant middle cerebral artery syndrome. Limitations for DC, such as the equipoise between immediate reduction in ICP and clinically meaningful functional outcomes, are discussed in support of future investigations.
Collapse
Affiliation(s)
- John K Yue
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Jonathan W Rick
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Hansen Deng
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Michael J Feldman
- Department of Neurological Surgery, Vanderbilt University, Nashville, TN, USA
| | - Ethan A Winkler
- Department of Neurological Surgery, University of California, San Francisco, CA, USA -
| |
Collapse
|
37
|
Hu HJ, Song M. Disrupted Ionic Homeostasis in Ischemic Stroke and New Therapeutic Targets. J Stroke Cerebrovasc Dis 2017; 26:2706-2719. [PMID: 29054733 DOI: 10.1016/j.jstrokecerebrovasdis.2017.09.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/30/2017] [Accepted: 09/06/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Stroke is a leading cause of long-term disability. All neuroprotectants targeting excitotoxicity have failed to become stroke medications. In order to explore and identify new therapeutic targets for stroke, we here reviewed present studies of ionic transporters and channels that are involved in ischemic brain damage. METHOD We surveyed recent literature from animal experiments and clinical reports in the databases of PubMed and Elsevier ScienceDirect to analyze ionic mechanisms underlying ischemic cell damage and suggest promising ideas for stroke therapy. RESULTS Dysfunction of ionic transporters and disrupted ionic homeostasis are most early changes that underlie ischemic brain injury, thus receiving sustained attention in translational stroke research. The Na+/K+-ATPase, Na+/Ca2+ Exchanger, ionotropic glutamate receptor, acid-sensing ion channels (ASICs), sulfonylurea receptor isoform 1 (SUR1)-regulated NCCa-ATP channels, and transient receptor potential (TRP) channels are critically involved in ischemia-induced cellular degenerating processes such as cytotoxic edema, excitotoxicity, necrosis, apoptosis, and autophagic cell death. Some ionic transporters/channels also act as signalosomes to regulate cell death signaling. For acute stroke treatment, glutamate-mediated excitotoxicity must be interfered within 2 hours after stroke. The SUR1-regulated NCCa-ATP channels, Na+/K+-ATPase, ASICs, and TRP channels have a much longer therapeutic window, providing new therapeutic targets for developing feasible pharmacological treatments toward acute ischemic stroke. CONCLUSION The next generation of stroke therapy can apply a polypharmacology strategy for which drugs are designed to target multiple ion transporters/channels or their interaction with neurotoxic signaling pathways. But a successful translation of neuroprotectants relies on in-depth analyses of cell death mechanisms and suitable animal models resembling human stroke.
Collapse
Affiliation(s)
- Hui-Jie Hu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingke Song
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
38
|
Targeting antioxidant enzyme expression as a therapeutic strategy for ischemic stroke. Neurochem Int 2016; 107:23-32. [PMID: 28043837 DOI: 10.1016/j.neuint.2016.12.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/05/2016] [Accepted: 12/17/2016] [Indexed: 11/24/2022]
Abstract
During ischemic stroke, neurons and glia are subjected to damage during the acute and neuroinflammatory phases of injury. Production of reactive oxygen species (ROS) from calcium dysregulation in neural cells and the invasion of activated immune cells are responsible for stroke-induced neurodegeneration. Scientists have failed thus far to identify antioxidant-based drugs that can enhance neural cell survival and improve recovery after stroke. However, several groups have demonstrated success in protecting against stroke by increasing expression of antioxidant enzymes in neural cells. These enzymes, which include but are not limited to enzymes in the glutathione peroxidase, catalase, and superoxide dismutase families, degrade ROS that otherwise damage cellular components such as DNA, proteins, and lipids. Several groups have identified cellular therapies including neural stem cells and human umbilical cord blood cells, which exert neuroprotective and oligoprotective effects through the release of pro-survival factors that activate PI3K/Akt signaling to upregulation of antioxidant enzymes. Other studies demonstrate that treatment with soluble factors released by these cells yield similar changes in enzyme expression after stroke. Treatment with the cytokine leukemia inhibitory factor increases the expression of peroxiredoxin IV and metallothionein III in glia and boosts expression of superoxide dismutase 3 in neurons. Through cell-specific upregulation of these enzymes, LIF and other Akt-inducing factors have the potential to protect multiple cell types against damage from ROS during the early and late phases of ischemic damage.
Collapse
|
39
|
Affiliation(s)
- Shao-Hua Yang
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Ran Liu
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
40
|
Wang H, Li P, Xu N, Zhu L, Cai M, Yu W, Gao Y. Paradigms and mechanisms of inhalational anesthetics mediated neuroprotection against cerebral ischemic stroke. Med Gas Res 2016; 6:194-205. [PMID: 28217291 PMCID: PMC5223310 DOI: 10.4103/2045-9912.196901] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cerebral ischemic stroke is a leading cause of serious long-term disability and cognitive dysfunction. The high mortality and disability of cerebral ischemic stroke is urging the health providers, including anesthesiologists and other perioperative professioners, to seek effective protective strategies, which are extremely limited, especially for those perioperative patients. Intriguingly, several commonly used inhalational anesthetics are recently suggested to possess neuroprotective effects against cerebral ischemia. This review introduces multiple paradigms of inhalational anesthetic treatments that have been investigated in the setting of cerebral ischemia, such as preconditioning, proconditioning and postconditioning with a variety of inhalational anesthetics. The pleiotropic mechanisms underlying these inhalational anesthetics-afforded neuroprotection against stroke are also discussed in detail, including the common pathways shared by most of the inhalational anesthetic paradigms, such as anti-excitotoxicity, anti-apoptosis and anti-inflammation. There are also distinct mechanisms involved in specific paradigms, such as preserving blood brain barrier integrity, regulating cerebral blood flow and catecholamine release. The ready availability of these inhalational anesthetics bedside and renders them a potentially translatable stroke therapy attracting great efforts for understanding of the underlying mechanisms.
Collapse
Affiliation(s)
- Hailian Wang
- Anesthesiology Department of Huashan Hospital, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China; Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peiying Li
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Na Xu
- Anesthesiology Department of Huashan Hospital, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Ling Zhu
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Mengfei Cai
- Anesthesiology Department of Huashan Hospital, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yanqin Gao
- Anesthesiology Department of Huashan Hospital, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China; Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
41
|
Wetterling F, Chatzikonstantinou E, Tritschler L, Meairs S, Fatar M, Schad LR, Ansar S. Investigating potentially salvageable penumbra tissue in an in vivo model of transient ischemic stroke using sodium, diffusion, and perfusion magnetic resonance imaging. BMC Neurosci 2016; 17:82. [PMID: 27927188 PMCID: PMC5143461 DOI: 10.1186/s12868-016-0316-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/25/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Diffusion magnetic resonance imaging (MRI) is the current-state-of-the-art technique to clinically investigate acute (0-24 h) ischemic stroke tissue. However, reduced apparent diffusion coefficient (ADC)-considered a marker of tissue damage-was observed to reverse spontaneously during the subacute stroke phase (24-72 h) which means that low ADC cannot be used to reflect the damaged tissue after 24 h in experimental and clinical studies. One reason for the change in ADC is that ADC values drop with cytotoxic edema (acute phase) and rise when vasogenic edema begins (subacute phase). Recently, combined 1H- and 23Na-MRI was proposed as a more accurate approach to improve delineation between reversible (penumbra) and irreversible ischemic injury (core). The aim of this study was to investigate the effects of reperfusion on the ADC and the sodium MRI signal after experimental ischemic stroke in rats in well-defined areas of different viability levels of the cerebral lesion, i.e. core and penumbra as defined via perfusion and histology. Transient middle cerebral artery occlusion was induced in male rats by using the intraluminal filament technique. MRI sodium, perfusion and diffusion measurement was recorded before reperfusion, shortly after reperfusion and 24 h after reperfusion. The animals were reperfused after 90 min of ischemia. RESULTS Sodium signal in core did not change before reperfusion, increased after reperfusion while sodium signal in penumbra was significantly reduced before reperfusion, but showed no changes after reperfusion compared to control. The ADC was significantly decreased in core tissue at all three time points compared to contralateral side. This decrease recovered above commonly applied viability thresholds in the core after 24 h. CONCLUSIONS Reduced sodium-MRI signal in conjunction with reduced ADC can serve as a viability marker for penumbra detection and complement hydrogen diffusion- and perfusion-MRI in order to facilitate time-independent assessment of tissue fate and cellular bioenergetics failure in stroke patients.
Collapse
Affiliation(s)
- Friedrich Wetterling
- Computer Assisted Clinical Medicine, Heidelberg University, Mannheim, Germany.,Trinity Institute of Neuroscience, University of Dublin, Dublin, Ireland
| | - Eva Chatzikonstantinou
- Department of Neurology, Universitätsmedizin Mannheim, Heidelberg University, Mannheim, Germany
| | - Laurent Tritschler
- Department of Neurology, Universitätsmedizin Mannheim, Heidelberg University, Mannheim, Germany.,CESP, INSERM UMRS1178, Faculté de Pharmacie, University Paris-Sud, Université Paris-Saclay, 92296, Chatenay-Malabry, France
| | - Stephen Meairs
- Department of Neurology, Universitätsmedizin Mannheim, Heidelberg University, Mannheim, Germany
| | - Marc Fatar
- Department of Neurology, Universitätsmedizin Mannheim, Heidelberg University, Mannheim, Germany
| | - Lothar R Schad
- Computer Assisted Clinical Medicine, Heidelberg University, Mannheim, Germany
| | - Saema Ansar
- Department of Neurology, Universitätsmedizin Mannheim, Heidelberg University, Mannheim, Germany. .,Division of Experimental Vascular Research, Department of Clinical Sciences, Lund University, Sölvegatan 17, BMC A13, 22184, Lund, Sweden.
| |
Collapse
|
42
|
Shi X, Doycheva DM, Xu L, Tang J, Yan M, Zhang JH. Sestrin2 induced by hypoxia inducible factor1 alpha protects the blood-brain barrier via inhibiting VEGF after severe hypoxic-ischemic injury in neonatal rats. Neurobiol Dis 2016; 95:111-21. [PMID: 27425892 DOI: 10.1016/j.nbd.2016.07.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 06/19/2016] [Accepted: 07/13/2016] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE Hypoxic ischemic (HI) encephalopathy remains the leading cause of perinatal brain injury resulting in long term disabilities. Stabilization of blood brain barrier (BBB) after HI is an important target, therefore, in this study we aim to determine the role of sestrin2, a stress inducible protein which is elevated after various insults, on BBB stabilization after moderate and severe HI injuries. METHODS Rat pups underwent common carotid artery ligation followed by either 150min (severe model) or 100min (moderate model) of hypoxia. 1h post HI, rats were intranasally administered with recombinant human sestrin2 (rh-sestrin2) and sacrificed for infarct area, brain water content, righting reflex and geotaxis reflex. Sestrin2 was silenced using siRNA and an activator/inhibitor of hypoxia inducible factor1α (HIF1α) was used to examine their roles on BBB permeability. RESULTS Rats subjected to severe HI exhibited larger infarct area and higher sestrin2 expression compared to rats in the moderate HI group. rh-sestrin2 attenuated brain infarct and edema, while silencing sestrin2 reversed these protective effects after severe HI. HIF1α induced sestrin2 activation in severe HI but not in moderate HI groups. A HIF1a agonist was shown to increase permeability of the BBB via vascular endothelial growth factor (VEGF) after moderate HI. However, after severe HI, HIF1α activated both VEGF and sestrin2. But HIF1α dependent sestrin2 activation was the predominant pathway after severe HI which inhibited VEGF and attenuated BBB permeability. CONCLUSIONS rh-sestrin2 attenuated BBB permeability via upregulation of endogenous sestrin2 which was induced by HIF1α after severe HI. However, HIF1α's effects as a prodeath or prosurvival signal were influenced by the severity of HI injury.
Collapse
Affiliation(s)
- Xudan Shi
- Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, China; Department of Anesthesiology and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Desislava Met Doycheva
- Department of Anesthesiology and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Liang Xu
- Department of Anesthesiology and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Jiping Tang
- Department of Anesthesiology and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Min Yan
- Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, China.
| | - John H Zhang
- Department of Anesthesiology and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| |
Collapse
|