1
|
Wipplinger C, Cattaneo A, Wipplinger TM, Lamllari K, Semmler F, Geske C, Messinger J, Nickl V, Beez A, Ernestus RI, Pham M, Westermaier T, Weiland J, Stetter C, Kunze E. Serum concentration-guided intravenous magnesium sulfate administration for neuroprotection in patients with aneurysmal subarachnoid hemorrhage: a retrospective evaluation of a 12-year single-center experience. Neurosurg Rev 2023; 46:256. [PMID: 37751032 PMCID: PMC10522732 DOI: 10.1007/s10143-023-02159-1] [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: 08/24/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/27/2023]
Abstract
Delayed cerebral infarction (DCI) is a major cause of morbidity and mortality in patients with aneurysmal subarachnoid hemorrhage (aSAH). The benefits of magnesium sulfate as an alternative treatment are controversial, and most previous studies examined its benefits only as adjunctive treatment to traditional nimodipine. We retrospectively analyzed aSAH patients records with magnesium sulfate between 2010 and 2021. We aimed for a serum magnesium concentration of 2-2.5 mmol/l between post-hemorrhage days 3 and 12. The patients were separated in three groups based on average serum magnesium concentration (magnesium >2 mmol/l, reduced magnesium 1.1-1.9 mmol/l, and no magnesium). Additionally, we assessed delayed cerebral infarction (DCI) and clinical outcome at follow-up, using the modified Rankin Scale (mRS), categorized in favorable (0-3) and unfavorable outcome (4-5). In this analysis, 548 patients were included. Hereof, radiological evidence of DCI could be found in 23.0% (n = 126) of patients. DCI rates were lower if patients' average serum magnesium was higher than 2 mmol/l (magnesium 18.8%, n = 85; reduced magnesium 38.3%, n = 23; no magnesium 51.4%, n = 18; p < 0.001). Also, at the last follow-up, patients in the group with a higher serum magnesium concentration had better outcome (favorable outcome: magnesium 64.7%, n = 293; reduced magnesium 50.0%, n = 30; no magnesium 34.3%, n = 12; p < 0.001). This 12-year study reveals the value of serum concentration-guided magnesium administration in aSAH patients. Our findings demonstrate the safety and efficacy when titrated to a serum concentration of 2-2.5 mmol/l. We observed higher rates of delayed cerebral infarction and unfavorable outcomes in patients with serum concentrations below 2 mmol/l.
Collapse
Affiliation(s)
- C Wipplinger
- Department of Neurosurgery, University Hospital of Würzburg, Würzburg, Germany.
| | - A Cattaneo
- Department of Neurosurgery, University Hospital of Würzburg, Würzburg, Germany
| | - T M Wipplinger
- Department of Neurosurgery, University Hospital of Würzburg, Würzburg, Germany
| | - K Lamllari
- Department of Neurosurgery, University Hospital of Würzburg, Würzburg, Germany
| | - F Semmler
- Department of Neurosurgery, University Hospital of Würzburg, Würzburg, Germany
| | - C Geske
- Department of Neurosurgery, University Hospital of Würzburg, Würzburg, Germany
| | - J Messinger
- Department of Neurosurgery, University Hospital of Würzburg, Würzburg, Germany
| | - V Nickl
- Department of Neurosurgery, University Hospital of Würzburg, Würzburg, Germany
| | - A Beez
- Department of Neurosurgery, University Hospital of Würzburg, Würzburg, Germany
| | - R-I Ernestus
- Department of Neurosurgery, University Hospital of Würzburg, Würzburg, Germany
| | - M Pham
- Department of Neuroradiology, University Hospital of Würzburg, Würzburg, Germany
| | - T Westermaier
- Department of Neurosurgery, Helios Amper-Klinikum Dachau, Dachau, Germany
| | - J Weiland
- Department of Neurosurgery, University Hospital of Würzburg, Würzburg, Germany
| | - C Stetter
- Department of Neurosurgery, University Hospital of Würzburg, Würzburg, Germany
| | - E Kunze
- Department of Neurosurgery, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
2
|
Okazaki K, Nakamura S, Koyano K, Konishi Y, Kondo M, Kusaka T. Neonatal asphyxia as an inflammatory disease: Reactive oxygen species and cytokines. Front Pediatr 2023; 11:1070743. [PMID: 36776908 PMCID: PMC9911547 DOI: 10.3389/fped.2023.1070743] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023] Open
Abstract
Neonatologists resuscitate asphyxiated neonates by every available means, including positive ventilation, oxygen therapy, and drugs. Asphyxiated neonates sometimes present symptoms that mimic those of inflammation, such as fever and edema. The main pathophysiology of the asphyxia is inflammation caused by hypoxic-ischemic reperfusion. At birth or in the perinatal period, neonates may suffer several, hypoxic insults, which can activate inflammatory cells and inflammatory mediator production leading to the release of larger quantities of reactive oxygen species (ROS). This in turn triggers the production of oxygen stress-induced high mobility group box-1 (HMGB-1), an endogenous damage-associated molecular patterns (DAMPs) protein bound to toll-like receptor (TLR) -4, which activates nuclear factor-kappa B (NF-κB), resulting in the production of excess inflammatory mediators. ROS and inflammatory mediators are produced not only in activated inflammatory cells but also in non-immune cells, such as endothelial cells. Hypothermia inhibits pro-inflammatory mediators. A combination therapy of hypothermia and medications, such as erythropoietin and melatonin, is attracting attention now. These medications have both anti-oxidant and anti-inflammatory effects. As the inflammatory response and oxidative stress play a critical role in the pathophysiology of neonatal asphyxia, these drugs may contribute to improving patient outcomes.
Collapse
Affiliation(s)
- Kaoru Okazaki
- Department of Neonatology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Shinji Nakamura
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kosuke Koyano
- Maternal Perinatal Center, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Yukihiko Konishi
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Masatoshi Kondo
- Department of Neonatology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Takashi Kusaka
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| |
Collapse
|
3
|
Cattaneo A, Wipplinger C, Geske C, Semmler F, Wipplinger TM, Griessenauer CJ, Weiland J, Beez A, Ernestus RI, Westermaier T, Kunze E, Stetter C. Investigating the relationship between high-dose norepinephrine administration and the incidence of delayed cerebral infarction in patients with aneurysmal subarachnoid hemorrhage: A single-center retrospective evaluation. PLoS One 2023; 18:e0283180. [PMID: 36943859 PMCID: PMC10030022 DOI: 10.1371/journal.pone.0283180] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/05/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND One of the longest-standing treatments to prevent delayed cerebral infarction (DCI) in patients with aneurysmal subarachnoid hemorrhage (aSAH) remains raising the blood pressure to a certain level of mean arterial pressure. This may require high doses of norepinephrine, which has been associated with severe end organ damage. With this study, we aimed to investigate the effects of norepinephrine on the incidence of DCI in a clinical setting. METHODS We conducted a retrospective evaluation of patients with aSAH admitted to our institution between November 2018 and March 2021. Potential risk factors for DCI were analyzed and significant predictors were assessed by means of a logistic regression analysis to account for potential confounders. RESULTS In this study, 104 patients were included. Hereof, 39 (38%) showed radiologic signs of DCI between day three and 14 post-intervention. These patients had more frequent vasospasms (n = 37 vs. 30, p = 0.022), a higher Hunt & Hess score (3 ± 2 vs. 2 ± 1, p = 0.004), a lower initial Glasgow Coma Scale score (9 ± 5 vs. 12 ± 4, p = 0.003) and received a higher median norepinephrine dose (20,356μg vs. 6,508μg, p < 0.001). A logistic regression analysis revealed that only high-dose norepinephrine administration (OR 2.84, CI 1.56-7.8) and vasospasm (OR 3.07, CI 1.2-7.84) appeared to be significant independent risk factors for DCI. CONCLUSION Our results indicate a significant association between higher dose norepinephrine administration and the occurrence of DCI. Future research including greater sample sizes and a prospective setting will be necessary to further investigate the relationship.
Collapse
Affiliation(s)
- Andrea Cattaneo
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | | | - Caroline Geske
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Florian Semmler
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Tamara M Wipplinger
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
- Department of Biobehavioral Sciences, Teachers College, Columbia University, New York, NY, United States of America
| | - Christoph J Griessenauer
- Department of Neurosurgery, Christian Doppler Klinik, Paracelsus Medical University, Salzburg, Austria
- Research Institute of Neurointervention, Paracelsus Medical University, Salzburg, Austria
| | - Judith Weiland
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Alexandra Beez
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Ralf-Ingo Ernestus
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Thomas Westermaier
- Department of Neurosurgery, Helios-Amper Klinikum Dachau, Dachau, Germany
| | - Ekkehard Kunze
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Christian Stetter
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
4
|
Mathew AA, Panonnummal R. A Mini Review on the Various Facets Effecting Brain Delivery of Magnesium and Its Role in Neurological Disorders. Biol Trace Elem Res 2022:10.1007/s12011-022-03517-8. [PMID: 36534337 DOI: 10.1007/s12011-022-03517-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022]
Abstract
Magnesium is an essential cation present in the body that participates in the regulation of various vital body functions. Maintaining normal level of magnesium is essential for proper brain functions by regulating the activities of numerous neurotransmitters and their receptors. Various studies have been reported that magnesium level is found to be declined in both neurological and psychiatric diseases. Declined magnesium level in the brain initiates various cumbersome effects like excitotoxicity, altered blood-brain permeability, oxidative stress, and inflammation, which may further worsen the disease condition. Shreds of evidence from the experimental and clinical studies proved that exogenous administration of magnesium is useful for correcting disease-induced alterations in the brain. But one of the major limiting factors in the use of magnesium for treatment purposes is its poor blood-brain barrier permeability. Various approaches like the administration of its organic salts as pidolate and threonate forms, and the combination with polyethylene glycol or mannitol have been tried to improve its permeability to make magnesium as a suitable drug for different neurological disorders. These results have shown their experimental efficacy in diseased animal models, but studies regarding the safety and efficacy in human subjects are currently underway. We present a comprehensive review on the role of magnesium in the maintenance of normal functioning of the brain and various approaches for improving its BBB permeability.
Collapse
Affiliation(s)
- Aparna Ann Mathew
- Amrita School of Pharmacy, Amrita Institute of Medical Science & Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Rajitha Panonnummal
- Amrita School of Pharmacy, Amrita Institute of Medical Science & Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India.
| |
Collapse
|
5
|
Ghozy S, Reda A, Varney J, Elhawary AS, Shah J, Murry K, Sobeeh MG, Nayak SS, Azzam AY, Brinjikji W, Kadirvel R, Kallmes DF. Neuroprotection in Acute Ischemic Stroke: A Battle Against the Biology of Nature. Front Neurol 2022; 13:870141. [PMID: 35711268 PMCID: PMC9195142 DOI: 10.3389/fneur.2022.870141] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/21/2022] [Indexed: 12/22/2022] Open
Abstract
Stroke is the second most common cause of global death following coronary artery disease. Time is crucial in managing stroke to reduce the rapidly progressing insult of the ischemic penumbra and the serious neurologic deficits that might follow it. Strokes are mainly either hemorrhagic or ischemic, with ischemic being the most common of all types of strokes. Thrombolytic therapy with recombinant tissue plasminogen activator and endovascular thrombectomy are the main types of management of acute ischemic stroke (AIS). In addition, there is a vital need for neuroprotection in the setting of AIS. Neuroprotective agents are important to investigate as they may reduce mortality, lessen disability, and improve quality of life after AIS. In our review, we will discuss the main types of management and the different modalities of neuroprotection, their mechanisms of action, and evidence of their effectiveness after ischemic stroke.
Collapse
Affiliation(s)
- Sherief Ghozy
- Department of Neuroradiology, Mayo Clinic, Rochester, MN, United States.,Nuffield Department of Primary Care Health Sciences and Department for Continuing Education (EBHC Program), Oxford University, Oxford, United Kingdom
| | - Abdullah Reda
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Joseph Varney
- School of Medicine, American University of the Caribbean, Philipsburg, Sint Maarten
| | | | - Jaffer Shah
- Medical Research Center, Kateb University, Kabul, Afghanistan
| | | | - Mohamed Gomaa Sobeeh
- Faculty of Physical Therapy, Sinai University, Cairo, Egypt.,Faculty of Physical Therapy, Cairo University, Giza, Egypt
| | - Sandeep S Nayak
- Department of Internal Medicine, NYC Health + Hospitals/Metropolitan, New York, NY, United States
| | - Ahmed Y Azzam
- Faculty of Medicine, October 6 University, Giza, Egypt
| | - Waleed Brinjikji
- Department of Neurosurgery, Mayo Clinic Rochester, Rochester, MN, United States
| | | | - David F Kallmes
- Department of Neuroradiology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
6
|
Current status and outlook of biodegradable metals in neuroscience and their potential applications as cerebral vascular stent materials. Bioact Mater 2021; 11:140-153. [PMID: 34938919 PMCID: PMC8665265 DOI: 10.1016/j.bioactmat.2021.09.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 09/01/2021] [Accepted: 09/18/2021] [Indexed: 12/12/2022] Open
Abstract
Over the past two decades, biodegradable metals (BMs) have emerged as promising materials to fabricate temporary biomedical devices, with the purpose of avoiding potential side effects of permanent implants. In this review, we first surveyed the current status of BMs in neuroscience, and briefly summarized the representative stents for treating vascular stenosis. Then, inspired by the convincing clinical evidence on the in vivo safety of Mg alloys as cardiovascular stents, we analyzed the possibility of producing biodegradable cerebrovascular Mg alloy stents for treating ischemic stroke. For these novel applications, some key factors should also be considered in designing BM brain stents, including the anatomic features of the cerebral vasculature, hemodynamic influences, neuro-cytocompatibility and selection of alloying elements. This work may provide insights into the future design and fabrication of BM neurological devices, especially for brain stents. The current status of the application of biodegradable metals (BM) in neuroscience was presented. We analyzed the possibility of producing biodegradable cerebrovascular Mg alloy stents for ischemic stroke treatment. Key factors in designing BM brain stents were discussed. This work may provide insights into the future design and fabrication of BM neurological devices, especially for brain stents.
Collapse
|
7
|
Weiland J, Beez A, Westermaier T, Kunze E, Sirén AL, Lilla N. Neuroprotective Strategies in Aneurysmal Subarachnoid Hemorrhage (aSAH). Int J Mol Sci 2021; 22:5442. [PMID: 34064048 PMCID: PMC8196706 DOI: 10.3390/ijms22115442] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/30/2021] [Accepted: 05/18/2021] [Indexed: 12/19/2022] Open
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) remains a disease with high mortality and morbidity. Since treating vasospasm has not inevitably led to an improvement in outcome, the actual emphasis is on finding neuroprotective therapies in the early phase following aSAH to prevent secondary brain injury in the later phase of disease. Within the early phase, neuroinflammation, thromboinflammation, disturbances in brain metabolism and early neuroprotective therapies directed against delayed cerebral ischemia (DCI) came into focus. Herein, the role of neuroinflammation, thromboinflammation and metabolism in aSAH is depicted. Potential neuroprotective strategies regarding neuroinflammation target microglia activation, metalloproteases, autophagy and the pathway via Toll-like receptor 4 (TLR4), high mobility group box 1 (HMGB1), NF-κB and finally the release of cytokines like TNFα or IL-1. Following the link to thromboinflammation, potential neuroprotective therapies try to target microthrombus formation, platelets and platelet receptors as well as clot clearance and immune cell infiltration. Potential neuroprotective strategies regarding metabolism try to re-balance the mismatch of energy need and supply following aSAH, for example, in restoring fuel to the TCA cycle or bypassing distinct energy pathways. Overall, this review addresses current neuroprotective strategies in aSAH, hopefully leading to future translational therapy options to prevent secondary brain injury.
Collapse
Affiliation(s)
- Judith Weiland
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
| | - Alexandra Beez
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
| | - Thomas Westermaier
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
- Department of Neurosurgery, Helios-Amper Klinikum Dachau, Krankenhausstr. 15, 85221 Dachau, Germany
| | - Ekkehard Kunze
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
| | - Anna-Leena Sirén
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
| | - Nadine Lilla
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
- Department of Neurosurgery, University Hospital Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| |
Collapse
|
8
|
Neuroprotective effect of magnesium supplementation on cerebral ischemic diseases. Life Sci 2021; 272:119257. [PMID: 33631176 DOI: 10.1016/j.lfs.2021.119257] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 01/31/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023]
Abstract
Ischemic encephalopathy is associated with a high mortality and rate of disability. The most common type of ischemic encephalopathy, ischemic stroke, is the second leading cause of death in the world. At present, the main treatment for ischemic stroke is to reopen blocked blood vessels. However, despite revascularization, many patients are not able to achieve good functional results. At the same time, the strict time window (<4.5 h) of thrombolytic therapy limits clinical application. Therefore, it is important to explore effective neuroprotective drugs for the treatment of ischemic stroke. Magnesium is a natural calcium antagonist, which exerts neuroprotective effects through various mechanisms. However, while most basic studies have shown that magnesium supplementation can help treat cerebral ischemia, intravenous magnesium supplementation in large clinical trials has failed to improve prognosis of ischemic patients. Therefore, we review the basic and clinical studies of magnesium supplementation for cerebral ischemia. According to the route of administration, treatment can be divided into intraperitoneal magnesium supplementation, intravenous magnesium supplementation, arterial magnesium supplementation and intracranial magnesium supplementation. We also summarized the potential influencing factors of magnesium ion intervention in cerebral ischemia injury. Finally, in combination with influencing factors derived from basic research, this article proposes three future research directions, including magnesium supplementation into the circulatory system combined with magnesium supplementation in the lateral ventricle, magnesium supplementation in the lateral ventricle combined with hypothermia therapy, and lateral ventricle magnesium supplementation combined with intracarotid magnesium supplementation combined with selective hypothermia.
Collapse
|
9
|
Mallah K, Couch C, Borucki DM, Toutonji A, Alshareef M, Tomlinson S. Anti-inflammatory and Neuroprotective Agents in Clinical Trials for CNS Disease and Injury: Where Do We Go From Here? Front Immunol 2020; 11:2021. [PMID: 33013859 PMCID: PMC7513624 DOI: 10.3389/fimmu.2020.02021] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Neurological disorders are major contributors to death and disability worldwide. The pathology of injuries and disease processes includes a cascade of events that often involve molecular and cellular components of the immune system and their interaction with cells and structures within the central nervous system. Because of this, there has been great interest in developing neuroprotective therapeutic approaches that target neuroinflammatory pathways. Several neuroprotective anti-inflammatory agents have been investigated in clinical trials for a variety of neurological diseases and injuries, but to date the results from the great majority of these trials has been disappointing. There nevertheless remains great interest in the development of neuroprotective strategies in this arena. With this in mind, the complement system is being increasingly discussed as an attractive therapeutic target for treating brain injury and neurodegenerative conditions, due to emerging data supporting a pivotal role for complement in promoting multiple downstream activities that promote neuroinflammation and degeneration. As we move forward in testing additional neuroprotective and immune-modulating agents, we believe it will be useful to review past trials and discuss potential factors that may have contributed to failure, which will assist with future agent selection and trial design, including for complement inhibitors. In this context, we also discuss inhibition of the complement system as a potential neuroprotective strategy for neuropathologies of the central nervous system.
Collapse
Affiliation(s)
- Khalil Mallah
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Christine Couch
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Health Sciences and Research, College of Health Professions, Medical University of South Carolina, Charleston, SC, United States
| | - Davis M. Borucki
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, United States
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC, United States
| | - Amer Toutonji
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, United States
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC, United States
| | - Mohammed Alshareef
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurological Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Ralph Johnson VA Medical Center, Charleston, SC, United States
| |
Collapse
|
10
|
Kim YS, Won YJ, Lim BG, Min TJ, Kim YH, Lee IO. Neuroprotective effects of magnesium L-threonate in a hypoxic zebrafish model. BMC Neurosci 2020; 21:29. [PMID: 32590943 PMCID: PMC7318545 DOI: 10.1186/s12868-020-00580-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 06/21/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Hypoxia inhibits the uptake of glutamate (a major neurotransmitter in the brain closely related to cognitive function) into brain cells, and the initial response of cells to cortical hypoxia depends on glutamate. Previous studies have suggested that magnesium may have protective effects against hypoxic injuries. In particular, magnesium L-threonate (MgT) may increase magnesium ion concentrations in the brain better than MgSO4 and improve cognitive function. METHODS We evaluated cell viability under hypoxic conditions in the MgT- and MgSO4-treated human SH-SY5Y neurons, in vivo behavior using the T-maze test following hypoxia in MgT-treated zebrafish, activity of brain mitochondrial dehydrogenase by 2,3,5-triphenyltetrazolium chloride (TTC) staining, and protein expression of the excitatory amino acid transporter (EAAT) 4 glutamate transporter by western blotting. RESULTS Among the groups treated with hypoxia, cell viability significantly increased when pre-treated with 1 or 10 mM MgT (p = 0.009 and 0.026, respectively). Despite hypoxic insult, MgT-treated zebrafish showed preferences for the red compartment (p = 0.025 for distance and p = 0.007 for frequency of entries), suggesting memory preservation. TTC staining showed reduced cerebral infarction and preserved absorbance in the MgT-treated zebrafish brain after hypoxia (p = 0.010 compared to the hypoxia group). In addition, western blot showed upregulation of EAAT4 protein in the MgT treated group. CONCLUSIONS Pre-treatment with MgT attenuated cell death and cerebral infarction due to hypoxia and protected cognitive function in zebrafish. In addition, MgT appeared to modulate expression of the glutamate transporter, EAAT4.
Collapse
Affiliation(s)
- Young-Sung Kim
- Department of Anesthesiology and Pain Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Young Ju Won
- Department of Anesthesiology and Pain Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Byung Gun Lim
- Department of Anesthesiology and Pain Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Too Jae Min
- Department of Anesthesiology and Pain Medicine, Korea University Ansan Hospital, Ansan, Korea
| | - Yeon-Hwa Kim
- Institute of Medical Science, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - Il Ok Lee
- Department of Anesthesiology and Pain Medicine, Korea University Guro Hospital, Seoul, Korea.
| |
Collapse
|
11
|
Schumacher SA, Toribio RE, Scansen B, Lakritz J, Bertone AL. Pharmacokinetics of magnesium and its effects on clinical variables following experimentally induced hypermagnesemia. J Vet Pharmacol Ther 2020; 43:577-590. [PMID: 32525571 DOI: 10.1111/jvp.12883] [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] [Received: 05/02/2019] [Revised: 03/22/2020] [Accepted: 05/07/2020] [Indexed: 12/16/2022]
Abstract
The objectives of this study were to describe pharmacokinetic and pharmacodynamic changes as a result of a single intravenous administration of magnesium sulfate (MgSO4 ) to healthy horses. MgSO4 is a magnesium salt that has been used to calm horses in equestrian competition and is difficult to regulate because magnesium is an essential constituent of all mammals. Six healthy adult female horses were administered a single intravenous dose of MgSO4 at 60 mg/kg of body weight over 5 min. Blood, urine, and cerebrospinal fluid (CSF) samples were collected, and cardiovascular parameters were monitored and echocardiograms performed at predetermined times. Noncompartmental pharmacokinetic analysis was applied to plasma concentrations of ionized magnesium (Mg2+ ). Objective data were analyzed using the Wilcoxon rank-sum test with p < .05 used as a determination for significance. Plasma concentrations of Mg2+ increased nearly fivefold, ionized calcium (Ca2+ ) decreased by nearly 10%, and the Ca2+ to Mg2+ ratio declined more than 3.5-fold and remained different than baseline until 24 hr (p < .05). Significant changes were seen with urinary fractional excretion of electrolytes, cardiovascular parameters, and echocardiographic measurements. No changes were detected in CSF electrolyte concentrations. The decrease in Ca2+ result of hypermagnesemia supports the interaction between these cations. Alterations detected in plasma electrolyte concentrations and urinary fractional excretion of electrolytes may serve as biomarkers for regulatory control for the nefarious administration of MgSO4 .
Collapse
Affiliation(s)
- Stephen A Schumacher
- College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA.,Department of Veterinary Clinical Sciences, The Ohio State University, Columbus, OH, USA
| | - Ramiro E Toribio
- College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA.,Department of Veterinary Clinical Sciences, The Ohio State University, Columbus, OH, USA
| | - Brian Scansen
- College of Veterinary Medicine & Biomedical Services, Colorado State University, Fort Collins, CO, USA
| | - Jeffrey Lakritz
- College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA.,Department of Veterinary Clinical Sciences, The Ohio State University, Columbus, OH, USA
| | - Alicia L Bertone
- College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA.,Department of Veterinary Clinical Sciences, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
12
|
Tinkov AA, Ajsuvakova OP, Skalny AV. A Case-Control Study of Essential and Toxic Trace Elements and Minerals in Hair of 0-4-Year-Old Children with Cerebral Palsy. Biol Trace Elem Res 2020; 195:399-408. [PMID: 31468294 DOI: 10.1007/s12011-019-01876-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 08/18/2019] [Indexed: 01/09/2023]
Abstract
The objective of the present study was to assess hair essential and toxic trace elements and minerals in children with cerebral palsy in relation to age of the examinees. A total of 70 children with cerebral palsy and 70 healthy controls aged 0-4 years old were enrolled in the present study. The examined children were also divided into two age groups of those younger and older than 2 years old. Hair trace element content was assessed using ICP-MS at NexION 300D (PerkinElmer, USA). The obtained data demonstrate that hair boron was more than 2-fold lower in CP children as compared with the control group. At the same time, hair Na, Se, and V levels were 21%, 12%, and 20% lower when compared with healthy controls, respectively. It is also notable that a 9% and 28% decrease in hair Fe and Li levels respectively were nearly significant. The observed alterations were more profound in a younger group of patients. No significant group difference in hair toxic metal and metalloid levels was observed between the general cohorts of children with and without CP. In regression models, only hair Al and Ca contents were significantly associated with the presence of cerebral palsy, whereas hair Mg, Na, Ni, and Se levels were characterized as significant negative predictors. The observed alteration in trace element metabolism may also provide an additional link between cerebral palsy, psychomotor delay, and certain diseases, including diabetes, epilepsy, and osteoporosis. However, further studies using other substrates (blood, urine) or biomarkers are required.
Collapse
Affiliation(s)
- Alexey A Tinkov
- Yaroslavl State University, Sovetskaya St., 14, Yaroslavl, Russia, 150003.
- IM Sechenov First Moscow State Medical University, Bolshaya Pirogovskaya St., 19-1, Moscow, Russia, 119146.
- RUDN University, Miklukho-Maklaya St., 6, Moscow, Russia, 117198.
| | - Olga P Ajsuvakova
- Yaroslavl State University, Sovetskaya St., 14, Yaroslavl, Russia, 150003
- IM Sechenov First Moscow State Medical University, Bolshaya Pirogovskaya St., 19-1, Moscow, Russia, 119146
- RUDN University, Miklukho-Maklaya St., 6, Moscow, Russia, 117198
| | - Anatoly V Skalny
- IM Sechenov First Moscow State Medical University, Bolshaya Pirogovskaya St., 19-1, Moscow, Russia, 119146
- RUDN University, Miklukho-Maklaya St., 6, Moscow, Russia, 117198
- Taipei Medical University, Wuxing St., 250, Taipei, 11031, Taiwan
| |
Collapse
|
13
|
Dhir N, Medhi B, Prakash A, Goyal MK, Modi M, Mohindra S. Pre-clinical to Clinical Translational Failures and Current Status of Clinical Trials in Stroke Therapy: A Brief Review. Curr Neuropharmacol 2020; 18:596-612. [PMID: 31934841 PMCID: PMC7457423 DOI: 10.2174/1570159x18666200114160844] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/31/2019] [Accepted: 12/28/2019] [Indexed: 12/16/2022] Open
Abstract
In stroke (cerebral ischemia), despite continuous efforts both at the experimental and clinical level, the only approved pharmacological treatment has been restricted to tissue plasminogen activator (tPA). Stroke is the leading cause of functional disability and mortality throughout worldwide. Its pathophysiology starts with energy pump failure, followed by complex signaling cascade that ultimately ends in neuronal cell death. Ischemic cascade involves excessive glutamate release followed by raised intracellular sodium and calcium influx along with free radicals' generation, activation of inflammatory cytokines, NO synthases, lipases, endonucleases and other apoptotic pathways leading to cell edema and death. At the pre-clinical stage, several agents have been tried and proven as an effective neuroprotectant in animal models of ischemia. However, these agents failed to show convincing results in terms of efficacy and safety when the trials were conducted in humans following stroke. This article highlights the various agents which have been tried in the past but failed to translate into stroke therapy along with key points that are responsible for the lagging of experimental success to translational failure in stroke treatment.
Collapse
Affiliation(s)
| | - Bikash Medhi
- Address correspondence to this author at the Department of Pharmacology, Research Block B, 4th Floor, Room no 4043, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India; E-mail:
| | | | | | | | | |
Collapse
|
14
|
Shen Y, Dai L, Tian H, Xu R, Li F, Li Z, Zhou J, Wang L, Dong J, Sun L. Treatment Of Magnesium-L-Threonate Elevates The Magnesium Level In The Cerebrospinal Fluid And Attenuates Motor Deficits And Dopamine Neuron Loss In A Mouse Model Of Parkinson's disease. Neuropsychiatr Dis Treat 2019; 15:3143-3153. [PMID: 31806980 PMCID: PMC6857673 DOI: 10.2147/ndt.s230688] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 10/28/2019] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Epidemiology research has demonstrated that magnesium (Mg) deficiency is associated with a high incidence of Parkinson's disease (PD). It is known that the systemic administration of MgSO4 is not able to elevate the Mg concentration in cerebrospinal fluid (CSF). This study aims to verify the protective effect of magnesium-L-threonate (MgT) in 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) mouse model. METHODS C57BL/6J mice were orally administered MgT or MgSO4 for 4 weeks, and received MPTP in the third week. After analysis of open-field and rotarod tests on the last day, tyrosine hydroxylase (TH) immunopositive cells and protein levels were quantified in the substantia nigra pars compacta (SNpc) and striatum. The expression of inducible nitric oxide synthase (iNOS) level was evaluated. Mg concentration in serum and CSF was measured after oral administration of MgSO4 or MgT in normal mice. Mg concentration in the CSF was increased in the mice treated with MgT but not MgSO4. RESULTS The total distance and mean speed in open-field tests, and the time spent on rotarod in the MgT group were increased, compared with MPTP group. The MgT treatment but not MgSO4 dose-dependently attenuated the loss of TH-positive neurons, and the reduction of the TH expression in the SNpc. The MgT treatment also inhibited the expression of iNOS as measured by immunohistochemistry and Western blots. Double-immunofluorescence staining of TH and iNOS showed iNOS-positive cells were collocalized for TH-positive cells. CONCLUSION The treatment with MgT is associated with an increase of Mg in the CSF. MgT, rather than MgSO4, can significantly attenuate MPTP-induced motor deficits and dopamine (DA) neuron loss.
Collapse
Affiliation(s)
- Yanling Shen
- Department of Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin541004, Guangxi, People’s Republic of China
- Department of Pathology, Affiliated Chenggong Hospital, Xiamen University, Xiamen, Fujian361000, People’s Republic of China
| | - Ling Dai
- Department of Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin541004, Guangxi, People’s Republic of China
| | - Haibo Tian
- Department of Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin541004, Guangxi, People’s Republic of China
- Department of Pathology, Fuling Central Hospital of Chongqing City, Chongqing408099, People’s Republic of China
| | - Runnan Xu
- Department of Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin541004, Guangxi, People’s Republic of China
| | - Fuying Li
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Zhuohang Li
- Department of Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin541004, Guangxi, People’s Republic of China
| | - Jeremy Zhou
- School of Pharmacy and Medical Sciences, and UniSA Cancer Research Institute, University of South Australia, Adelaide, SA5001, Australia
| | - Liping Wang
- School of Pharmacy and Medical Sciences, and UniSA Cancer Research Institute, University of South Australia, Adelaide, SA5001, Australia
| | - Jianghui Dong
- Department of Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin541004, Guangxi, People’s Republic of China
- School of Pharmacy and Medical Sciences, and UniSA Cancer Research Institute, University of South Australia, Adelaide, SA5001, Australia
| | - Liyuan Sun
- Department of Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin541004, Guangxi, People’s Republic of China
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| |
Collapse
|
15
|
Jafari M, Di Napoli M, Lattanzi S, Mayer SA, Bachour S, Bershad EM, Damani R, Datta YH, Divani AA. Serum magnesium level and hematoma expansion in patients with intracerebral hemorrhage. J Neurol Sci 2019; 398:39-44. [PMID: 30682519 DOI: 10.1016/j.jns.2019.01.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/25/2018] [Accepted: 01/15/2019] [Indexed: 12/28/2022]
Abstract
Spontaneous intracerebral hemorrhage (ICH) is a devastating subtype of stroke that results in significant rates of mortality and morbidities. The initial hematoma volume, hematoma expansion (HE), blood pressure (BP), and coagulopathy are considered strong predictors of clinical outcomes and mortality. Low serum magnesium (Mg++) levels have been shown to be associated with larger initial hematoma and greater HE. Coagulopathy, platelet dysfunction, high BP, and increased inflammatory response might form the mechanistic link between low serum Mg++ levels, larger hematoma size and greater HE. However, randomized clinical trials administering intravenous Mg++ have shown no benefit over placebo in ICH patients. The confounding effect of hypocalcemia and a delay in Mg++ trafficking across the blood-brain barrier might explain the futile results for intravenous Mg++ therapy. In the current review, we will discuss the evidence regarding the possible role of low serum Mg++ level on HE in acute ICH.
Collapse
Affiliation(s)
- Mostafa Jafari
- Department of Neurology, University of Minnesota, Minneapolis, MN, United States
| | - Mario Di Napoli
- Department of Neurology, San Camillo de' Lellis District General Hospital, Rieti, Italy
| | - Simona Lattanzi
- Neurological Clinic, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Stephan A Mayer
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| | - Salam Bachour
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, United States
| | - Eric M Bershad
- Department of Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Rahul Damani
- Department of Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Yvonne H Datta
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Afshin A Divani
- Department of Neurology, University of Minnesota, Minneapolis, MN, United States; Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
16
|
Chang JJ, Armonda R, Goyal N, Arthur AS. Magnesium: Pathophysiological mechanisms and potential therapeutic roles in intracerebral hemorrhage. Neural Regen Res 2019; 14:1116-1121. [PMID: 30804233 PMCID: PMC6425828 DOI: 10.4103/1673-5374.251189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Intracerebral hemorrhage (ICH) remains the second-most common form of stroke with high morbidity and mortality. ICH can be divided into two pathophysiological stages: an acute primary phase, including hematoma volume expansion, and a subacute secondary phase consisting of blood-brain barrier disruption and perihematomal edema expansion. To date, all major trials for ICH have targeted the primary phase with therapies designed to reduce hematoma expansion through blood pressure control, surgical evacuation, and hemostasis. However, none of these trials has resulted in improved clinical outcomes. Magnesium is a ubiquitous element that also plays roles in vasodilation, hemostasis, and blood-brain barrier preservation. Animal models have highlighted potential therapeutic roles for magnesium in neurological diseases specifically targeting these pathophysiological mechanisms. Retrospective studies have also demonstrated inverse associations between admission magnesium levels and hematoma volume, hematoma expansion, and clinical outcome in patients with ICH. These associations, coupled with the multifactorial role of magnesium that targets both primary and secondary phases of ICH, suggest that magnesium may be a viable target of study in future ICH studies.
Collapse
Affiliation(s)
- Jason J Chang
- Department of Critical Care Medicine, MedStar Washington Hospital Center; Department of Neurology, Georgetown University School of Medicine, Washington, DC, USA
| | - Rocco Armonda
- Department of Neurosurgery, Georgetown University School of Medicine, Washington, DC, USA
| | - Nitin Goyal
- Department of Neurology, University of Tennessee Health Science Center; Semmes Murphey Clinic, Memphis, TN, USA
| | - Adam S Arthur
- Semmes Murphey Clinic; Department of Neurosurgery, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
17
|
Goenka L, Uppugunduri Satyanarayana CR, S SK, George M. Neuroprotective agents in Acute Ischemic Stroke-A Reality Check. Biomed Pharmacother 2018; 109:2539-2547. [PMID: 30551514 DOI: 10.1016/j.biopha.2018.11.041] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 10/22/2018] [Accepted: 11/10/2018] [Indexed: 11/29/2022] Open
Affiliation(s)
- Luxitaa Goenka
- Department of Clinical Pharmacology, SRM Medical College Hospital & Research Centre, Kattankulathur, Chennai, Tamil Nadu, 603203 India
| | - Chakradhara Rao Uppugunduri Satyanarayana
- Platform of Pediatric Onco-Hematology (CANSEARCH Laboratory), Department of Pediatrics, University of Geneva, Bâtiment Tulipe, Avenue De La Roseraie, 641205 Geneva, Switzerland
| | - Suresh Kumar S
- Department of Pharmacology, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, Ras Al Khaymah, United Arab Emirates
| | - Melvin George
- Department of Clinical Pharmacology, SRM Medical College Hospital & Research Centre, Kattankulathur, Chennai, Tamil Nadu, 603203 India.
| |
Collapse
|
18
|
Lee TM, Chang NC, Lin SZ. Effect of proton pump inhibitors on sympathetic hyperinnervation in infarcted rats: Role of magnesium. PLoS One 2018; 13:e0202979. [PMID: 30153299 PMCID: PMC6112652 DOI: 10.1371/journal.pone.0202979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/13/2018] [Indexed: 11/30/2022] Open
Abstract
The long-term use of proton pump inhibitors (PPIs) has been shown to increase the risk of cardiovascular mortality, however the molecular mechanisms are unknown. Superoxide has been implicated in the regulation of nerve growth factor (NGF), a mediator of sympathetic innervation. The purpose of this study was to determine whether PPIs increase ventricular arrhythmias through magnesium-mediated superoxide production in infarcted rats. Male Wistar rats were randomly assigned to receive vehicle, omeprazole, omeprazole + magnesium sulfate, or famotidine treatment for 4 weeks starting 24 hours after the induction of myocardial infarction by ligating the coronary artery. Increased myocardial superoxide and nitrotyrosine levels were noted post-infarction, in addition to a significant upregulation of NGF expression on mRNA and protein levels. Sympathetic hyperinnervation after infarction was confirmed by measuring myocardial norepinephrine and immunofluorescent analysis. Compared with the vehicle, omeprazole-treated infarcted rats had significantly reduced myocardial magnesium content, increased oxidant production, and increased sympathetic innervation, which in turn increased ventricular arrhythmias. These effects were prevented by the coadministration of magnesium sulfate. In an in vivo study, an omeprazole-induced increase in NGF was associated with a superoxide pathway, which was further confirmed by an ex vivo study showing the attenuation of NGF levels after coadministration of the superoxide scavenger Tiron. Magnesium sulfate did not further attenuate NGF levels compared with omeprazole + Tiron. Our results indicate that the long-term administration of PPIs was associated with reduced tissue magnesium content and increased myocardial superoxide production, which exacerbated ventricular arrhythmias after infarction. Magnesium may be a potential target for PPI-related arrhythmias after infarction.
Collapse
Affiliation(s)
- Tsung-Ming Lee
- Cardiovascular Institute, An Nan Hospital, China Medical University, Tainan, Taiwan
- Department of Medicine, China Medical University, Taichung, Taiwan
- Department of Internal Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Nen-Chung Chang
- Department of Internal Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Shinn-Zong Lin
- Department of Neurosurgery, Buddhist Tzu Chi General hospital, Tzu Chi University, Hualien, Taiwan
- * E-mail:
| |
Collapse
|
19
|
Rimkute L, Kraujalis T, Snipas M, Palacios-Prado N, Jotautis V, Skeberdis VA, Bukauskas FF. Modulation of Connexin-36 Gap Junction Channels by Intracellular pH and Magnesium Ions. Front Physiol 2018; 9:362. [PMID: 29706896 PMCID: PMC5906587 DOI: 10.3389/fphys.2018.00362] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 03/23/2018] [Indexed: 11/13/2022] Open
Abstract
Connexin-36 (Cx36) protein forms gap junction (GJ) channels in pancreatic beta cells and is also the main Cx isoform forming electrical synapses in the adult mammalian brain. Cx36 GJs can be regulated by intracellular pH (pHi) and cytosolic magnesium ion concentration ([Mg2+]i), which can vary significantly under various physiological and pathological conditions. However, the combined effect and relationship of these two factors over Cx36-dependent coupling have not been previously studied in detail. Our experimental results in HeLa cells expressing Cx36 show that changes in both pHi and [Mg2+]i affect junctional conductance (gj) in an interdependent manner; in other words, intracellular acidification cause increase or decay in gj depending on whether [Mg2+]i is high or low, respectively, and intracellular alkalization cause reduction in gj independently of [Mg2+]i. Our experimental and modelling data support the hypothesis that Cx36 GJ channels contain two separate gating mechanisms, and both are differentially sensitive to changes in pHi and [Mg2+]i. Using recombinant Cx36 we found that two glutamate residues in the N-terminus could be partly responsible for the observed interrelated effect of pHi and [Mg2+]i. Mutation of glutamate at position 8 attenuated the stimulatory effect of intracellular acidification at high [Mg2+]i, while mutation at position 12 and double mutation at both positions reversed stimulatory effect to inhibition. Moreover, Cx36*E8Q lost the initial increase of gj at low [Mg2+]i and double mutation lost the sensitivity to high [Mg2+]i. These results suggest that E8 and E12 are involved in regulation of Cx36 GJ channels by Mg2+ and H+ ions.
Collapse
Affiliation(s)
- Lina Rimkute
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Tadas Kraujalis
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Applied Informatics, Kaunas University of Technology, Kaunas, Lithuania
| | - Mindaugas Snipas
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Mathematical Modelling, Kaunas University of Technology, Kaunas, Lithuania
| | - Nicolas Palacios-Prado
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Vaidas Jotautis
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Vytenis A. Skeberdis
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | |
Collapse
|
20
|
Wang X, An F, Wang S, An Z, Wang S. Orientin Attenuates Cerebral Ischemia/Reperfusion Injury in Rat Model through the AQP-4 and TLR4/NF-κB/TNF-α Signaling Pathway. J Stroke Cerebrovasc Dis 2017. [PMID: 28645524 DOI: 10.1016/j.jstrokecerebrovasdis.2017.05.002] [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] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Orientin has been reported to have extensive pharmaceutical effects of antioxidant, anti-inflammatory, antithrombosis, antiapoptosis, and so on. In the present study, we tried to investigate the protective effects of orientin on cerebral ischemia-reperfusion (I/R) injury and explored the possible mechanisms. METHODS Middle cerebral artery occlusion rat model was established and then treated with low, middle, and high concentrations of orientin, respectively, with edaravone as a positive control. The treatment effect of orientin was evaluated by measuring the neurological deficit score, cerebral infarction, brain edema, oxidative stress, excitatory amino acids release, the expression levels of aquaporin-4 (AQP-4), and related inflammatory molecules using different methods including immunohistochemistry, enzyme-linked immunosorbent assay, real-time PCR, and western blot. Moreover, morphological and structural changes were also observed by hematoxylin-eosin staining and transmission electron microscope. RESULTS Orientin provided a significant reduction on neurological deficits, cerebral infarction, cerebral edema, oxidative damage, and neurotoxicity of excitatory amino acids compared to model group (P < .05) in a dose-dependent manner. In addition, orientin substantially downregulated AQP-4 and inflammatory factors expression (P < .05) and improved cell morphology and structure in rats following I/R injury. CONCLUSION Orientin was able to mediate noticeable protection against cerebral I/R injury through the attenuation of oxidative stress and neurotoxicity of amino acids and inhibiting the upregulation of AQP-4 and inflammatory cytokines.
Collapse
Affiliation(s)
- Xiaoru Wang
- Department of Pharmacy, Hebei North University, Zhangjiakou, Hebei Province, China
| | - Fang An
- Graduate Faculty, Hebei North University, Zhangjiakou, Hebei Province, China
| | - Shulin Wang
- Department of Pharmacy, Hebei North University, Zhangjiakou, Hebei Province, China
| | - Zexin An
- Department of Information, First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, China
| | - Shuhua Wang
- Department of Pharmacy, Hebei North University, Zhangjiakou, Hebei Province, China.
| |
Collapse
|
21
|
Christophe BR, Mehta SH, Garton ALA, Sisti J, Connolly ES. Current and future perspectives on the treatment of cerebral ischemia. Expert Opin Pharmacother 2017; 18:573-580. [PMID: 28393614 DOI: 10.1080/14656566.2017.1309022] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION After heart disease and combined forms of cancer, stroke is the leading cause of death in the United States. Currently, tissue-plasminogen activator (tPA) thrombolysis is the only thrombolytic therapy that has been shown to improve patient outcome. Presently, the only antithrombotic drug treatment that has proven effective at improving acute ischemic stroke patient outcome is aspirin administration. Despite these studies, no clinical trials have yet demonstrated a reliably effective pharmacological treatment. Areas covered: We conducted a search of recent drug studies for ischemic stroke on clinicaltrials.gov in addition to a literature search for acute ischemic stroke therapy using PubMed. This review details our findings of recent advancements in the pharmacological treatment of acute ischemic stroke. Expert commentary: We concluded that recent attempts to establish new pharmacological treatment protocols for acute ischemic stroke have had limited success, but many Phase III and Phase IV clinical trials demonstrate promise. Moreover, several studies have demonstrated the efficacy of dual-antiplatelet therapies at reducing risk of secondary stroke. Studies for novel therapeutic targets for neuroprotection have been largely unsuccessful. Some trials had positive results; however, there is much room for improvement and other studies show promise in their preliminary stages.
Collapse
Affiliation(s)
- Brandon R Christophe
- a Department of Neurological Surgery , Columbia University Medical Center , New York , NY , USA
| | - Shyle H Mehta
- a Department of Neurological Surgery , Columbia University Medical Center , New York , NY , USA
| | - Andrew L A Garton
- a Department of Neurological Surgery , Columbia University Medical Center , New York , NY , USA
| | - Jonathan Sisti
- a Department of Neurological Surgery , Columbia University Medical Center , New York , NY , USA
| | - E Sander Connolly
- a Department of Neurological Surgery , Columbia University Medical Center , New York , NY , USA
| |
Collapse
|
22
|
Khalessi N, Mazouri A, Bassirnia M, Afsharkhas L. Comparison between serum magnesium levels of asphyxiated neonates and normal cases. Med J Islam Repub Iran 2017; 31:19. [PMID: 28955669 PMCID: PMC5609322 DOI: 10.18869/mjiri.31.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Indexed: 11/25/2022] Open
Abstract
Background: Asphyxia is one the most important causes of neonatal mortality and morbidity. It is suggested that magnesium may have a protective role against cellular damage during hypoxic brain insult, or change effect post-asphyxia consequences. Our study was performed for comparison of serum magnesium in neonates with and without asphyxia. Methods: This study was done in Neonatal Ward of Ali-Asghar hospital, from January 2010 to 2011 in Tehran, Iran. Serum magnesium levels of seventy-six cases with a diagnosis of asphyxia grade 2 were compared with 76 normal newborns. Collected data including gestational age, sex, birth weight and serum magnesium levels were analyzed by SPSS software. Results: Of 152 neonates, 81 (53.3%) were male. Mean gestational age was 37.9 ± 1.07 weeks. Mean birth weights were 3172.9± 411.20 grams. Mean serum magnesium levels were compared in asphyxiated and normal neonates and between two groups significant difference was found (p=0.01). The odds ratio was 2.188 (with lower1.826, upper 2.626 and confidence interval 95 percent) which suggested a significant correlation between asphyxia and hypomagnesemia. Conclusion: This study showed that serum magnesium levels in neonates with asphyxia was significantly lower than normal neonates and asphyxia can lead to hypomagnesemia.
Collapse
Affiliation(s)
- Nasrin Khalessi
- Ali-Asghar Children’s Hospital, Iran University of Medical Science, Tehran, Iran
| | | | - Mandana Bassirnia
- Ali-Asghar Children’s Hospital, Iran University of Medical Science, Tehran, Iran
| | - Ladan Afsharkhas
- Ali-Asghar Children’s Hospital, Iran University of Medical Science, Tehran, Iran
| |
Collapse
|
23
|
Dal Ben M, Bottin C, Zanconati F, Tiribelli C, Gazzin S. Evaluation of region selective bilirubin-induced brain damage as a basis for a pharmacological treatment. Sci Rep 2017; 7:41032. [PMID: 28102362 PMCID: PMC5244479 DOI: 10.1038/srep41032] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/14/2016] [Indexed: 12/20/2022] Open
Abstract
The neurologic manifestations of neonatal hyperbilirubinemia in the central nervous system (CNS) exhibit high variations in the severity and appearance of motor, auditory and cognitive symptoms, which is suggestive of a still unexplained selective topography of bilirubin-induced damage. By applying the organotypic brain culture (OBC: preserving in vitro the cellular complexity, connection and architecture of the in vivo brain) technique to study hyperbilirubinemia, we mapped the regional target of bilirubin-induced damage, demonstrated a multifactorial toxic action of bilirubin, and used this information to evaluate the efficacy of drugs applicable to newborns to protect the brain. OBCs from 8-day-old rat pups showed a 2–13 fold higher sensitivity to bilirubin damage than 2-day-old preparations. The hippocampus, inferior colliculus and cerebral cortex were the only brain regions affected, presenting a mixed inflammatory-oxidative mechanism. Glutamate excitotoxicity was appreciable in only the hippocampus and inferior colliculus. Single drug treatment (indomethacin, curcumin, MgCl2) significantly improved cell viability in all regions, while the combined (cocktail) administration of the three drugs almost completely prevented damage in the most affected area (hippocampus). Our data may supports an innovative (complementary to phototherapy) approach for directly protecting the newborn brain from bilirubin neurotoxicity.
Collapse
Affiliation(s)
- Matteo Dal Ben
- Italian Liver Foundation (Fondazione Italiana Fegato), AREA Science Park, Trieste, Italy
| | - Cristina Bottin
- Department of Medical Sciences (Dipartimento di Scienze Mediche), Ospedale di Cattinara, Univestità degli Studi di Trieste, Trieste, Italy
| | - Fabrizio Zanconati
- Department of Medical Sciences (Dipartimento di Scienze Mediche), Ospedale di Cattinara, Univestità degli Studi di Trieste, Trieste, Italy
| | - Claudio Tiribelli
- Italian Liver Foundation (Fondazione Italiana Fegato), AREA Science Park, Trieste, Italy
| | - Silvia Gazzin
- Italian Liver Foundation (Fondazione Italiana Fegato), AREA Science Park, Trieste, Italy
| |
Collapse
|
24
|
|
25
|
Itoh K, Maki T, Shindo A, Egawa N, Liang AC, Itoh N, Lo EH, Lok J, Arai K. Magnesium sulfate protects oligodendrocyte lineage cells in a rat cell-culture model of hypoxic-ischemic injury. Neurosci Res 2015; 106:66-9. [PMID: 26699082 DOI: 10.1016/j.neures.2015.12.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 12/01/2015] [Accepted: 12/04/2015] [Indexed: 01/02/2023]
Abstract
Hypoxic-ischemic (HI) brain injury in newborns results in serious damage. Magnesium sulfate has been clinically used as a cyto-protective agent against HI brain injury in newborns in some countries, including Japan. However, it is not clear how magnesium exerts this effect and how it acts on the individual types of cells within the newborn brain. In this study, we exposed cultured rat oligodendrocyte precursor cells to magnesium sulfate during the period when they differentiate into oligodendrocytes, and showed that magnesium-exposed oligodendrocytes exhibited more resistance to HI injury. Our data may support the use of magnesium sulfate in the clinical setting.
Collapse
Affiliation(s)
- Kanako Itoh
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA; Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Takakuni Maki
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Akihiro Shindo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Naohiro Egawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Anna C Liang
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Naoki Itoh
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Josephine Lok
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA; Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
26
|
|
27
|
Titomanlio L, Fernández-López D, Manganozzi L, Moretti R, Vexler ZS, Gressens P. Pathophysiology and neuroprotection of global and focal perinatal brain injury: lessons from animal models. Pediatr Neurol 2015; 52:566-584. [PMID: 26002050 PMCID: PMC4720385 DOI: 10.1016/j.pediatrneurol.2015.01.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 01/16/2015] [Accepted: 01/24/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Arterial ischemic stroke occurs more frequently in term newborns than in the elderly, and brain immaturity affects mechanisms of ischemic injury and recovery. The susceptibility to injury of the brain was assumed to be lower in the perinatal period as compared with childhood. This concept was recently challenged by clinical studies showing marked motor disabilities after stroke in neonates, with the severity of motor and cortical sensory deficits similar in both perinatal and childhood ischemic stroke. Our understanding of the triggers and the pathophysiological mechanisms of perinatal stroke has greatly improved in recent years, but many factors remain incompletely understood. METHODS In this review, we focus on the pathophysiology of perinatal stroke and on therapeutic strategies that can protect the immature brain from the consequences of stroke by targeting inflammation and brain microenvironment. RESULTS Studies in neonatal rodent models of cerebral ischemia have suggested a potential role for soluble inflammatory molecules as important modulators of injury and recovery. A great effort is underway to investigate neuroprotective molecules based on our increasing understanding of the pathophysiology. CONCLUSION In this review, we provide a comprehensive summary of new insights concerning pathophysiology of focal and global perinatal brain injury and their implications for new therapeutic approaches.
Collapse
Affiliation(s)
- Luigi Titomanlio
- Pediatric Emergency Department, APHP, Robert Debré Hospital, Paris, France
- Inserm, U1141, F-75019 Paris, France
| | - David Fernández-López
- Department of Neurology, University of California San Francisco, San Francisco, CA, 94158-0663, USA
| | - Lucilla Manganozzi
- Pediatric Emergency Department, APHP, Robert Debré Hospital, Paris, France
- Inserm, U1141, F-75019 Paris, France
| | | | - Zinaida S. Vexler
- Department of Neurology, University of California San Francisco, San Francisco, CA, 94158-0663, USA
| | - Pierre Gressens
- Inserm, U1141, F-75019 Paris, France
- Univ Paris Diderot, Sorbonne Paris Cité, UMRS 676, F-75019 Paris, France
- PremUP, Paris, France
- Centre for the Developing Brain, King’s College, St Thomas’ Campus, London SE1 7EH, UK
| |
Collapse
|
28
|
Galinsky R, Bennet L, Groenendaal F, Lear CA, Tan S, van Bel F, Juul SE, Robertson NJ, Mallard C, Gunn AJ. Magnesium is not consistently neuroprotective for perinatal hypoxia-ischemia in term-equivalent models in preclinical studies: a systematic review. Dev Neurosci 2015; 36:73-82. [PMID: 24854050 DOI: 10.1159/000362206] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 03/13/2014] [Indexed: 11/19/2022] Open
Abstract
There is an important unmet need to further improve the outcome of neonatal encephalopathy in term infants. Meta-analyses of large controlled trials now suggest that maternal magnesium sulfate (MgSO4) therapy is associated with a reduced risk of cerebral palsy and gross motor dysfunction after premature birth, but that it has no effect on death or disability. Because of this inconsistency, it remains controversial whether MgSO4 is clinically neuroprotective and, thus, it is unclear whether it would be appropriate to test MgSO4 for treatment of encephalopathy in term infants. We therefore systematically reviewed the preclinical evidence for neuroprotection with MgSO4 before or after hypoxic-ischemic encephalopathy (HIE) in term-equivalent perinatal and adult animals. The outcomes were highly inconsistent between studies. Although there were differences in dose and timing of administration, there was evidence that beneficial effects of MgSO4 were associated with confounding mild hypothermia and, strikingly, the studies that included rigorous maintenance of environmental temperature or body temperature consistently suggested a lack of effect. On balance, these preclinical studies suggest that peripherally administered MgSO4 is unlikely to be neuroprotective. Rigorous testing in translational animal models of perinatal HIE is needed before MgSO4 should be considered in clinical trials for encephalopathy in term infants.
Collapse
|
29
|
Neuroprotection for ischaemic stroke: Current status and challenges. Pharmacol Ther 2015; 146:23-34. [DOI: 10.1016/j.pharmthera.2014.09.003] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 09/02/2014] [Indexed: 12/31/2022]
|
30
|
The use of intravenous magnesium in non-preeclamptic pregnant women: fetal/neonatal neuroprotection. Arch Gynecol Obstet 2014; 291:969-75. [DOI: 10.1007/s00404-014-3581-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 12/05/2014] [Indexed: 10/24/2022]
|
31
|
Long-term outcomes after immediate aortic repair for acute type A aortic dissection complicated by coma. J Thorac Cardiovasc Surg 2014; 148:1013-8; discussion 1018-9. [DOI: 10.1016/j.jtcvs.2014.06.053] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/26/2014] [Accepted: 06/30/2014] [Indexed: 11/30/2022]
|
32
|
Yperzeele L, Van Hooff RJ, De Smedt A, Valenzuela Espinoza A, Van de Casseye R, Hubloue I, De Keyser J, Brouns R. Prehospital stroke care: limitations of current interventions and focus on new developments. Cerebrovasc Dis 2014; 38:1-9. [PMID: 25116305 DOI: 10.1159/000363617] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 05/15/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The global burden of stroke is immense, both in medical and economic terms. With the aging population and the ongoing industrialization of the third world, stroke prevalence is expected to increase and will have a major effect on national health expenditures. Currently, the medical treatment for acute ischemic stroke is limited to intravenous recombinant tissue plasminogen activator (IV r-tPA), but its time dependency leads to low utilization rates in routine clinical practice. Prehospital delay contributes significantly to delayed or missed treatment opportunities in acute stroke. State-of-the-art acute stroke care, starting in the prehospital phase, could thereby reduce the disease burden and its enormous financial costs. SUMMARY The first part of this review focuses on current education measures for the general public, the emergency medical services (EMS) dispatchers and paramedics. Although much has been expected of these measures to improve stroke care, no major effects on prehospital delay or missed treatment opportunities have been demonstrated over the years. Most interventional studies showed little or no effect on the onset-to-door time, IV r-tPA utilization rates or outcome, except for prenotification of the receiving hospital by the EMS. No data are currently available on the cost-effectiveness of these commonly used measures. In the second part, we discuss new developments for the improvement of prehospital stroke diagnosis and treatment which could open new perspectives in the nearby future. These include the implementation of prehospital telestroke and the deployment of mobile stroke units. These approaches may improve patient care and could serve as a platform for prehospital clinical trials. Other opportunities include the implementation of noninvasive diagnostics (like transcranial ultrasound and blood-borne biomarkers) and the reevaluation of neuroprotective strategies in the prehospital phase. Key Messages: Timely initiation of treatment can effectively reduce the medical and economic burden of stroke and should begin with optimal prehospital stroke care. For this, prehospital telemedicine is a particularly attractive approach because it is a scalable solution that has the potential to rapidly optimize acute stroke care at limited cost.
Collapse
Affiliation(s)
- Laetitia Yperzeele
- Department of Neurology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|