1
|
Sasongko AB, Perdana Wahjoepramono PO, Halim D, Aviani JK, Adam A, Tsai YT, Wahjoepramono EJ, July J, Achmad TH. Potential blood biomarkers that can be used as prognosticators of spontaneous intracerebral hemorrhage: A systematic review and meta-analysis. PLoS One 2025; 20:e0315333. [PMID: 39970158 PMCID: PMC11838903 DOI: 10.1371/journal.pone.0315333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 11/22/2024] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Predicting nontraumatic spontaneous intracerebral hemorrhage (SICH) patient prognosis has been commonly practiced, particularly when providing informed consent and considering surgical treatment. Biomarkers might provide more real-time evaluation of SICH patients' condition than clinical prognostic scoring systems. This study aimed to evaluate the reliability of blood biomarkers in predicting prognosis in SICH patients by systematic review and meta-analysis. METHODS Studies that evaluated the association of blood biomarker(s) with mortality and/or functional outcome in SICH patients up to October 11, 2024, were identified through PubMed, Google Scholars, Scopus databases, and reference lists. Studies that satisfied the inclusion criteria were included in the meta-analyses. Good functional outcome was defined by patient's Glasgow Outcome Scale (GOS) ≥ 4 or modified Rankin scale mRS ≤ 2. Blood biomarkers were classified into the following categories: angiogenic factors, growth factors, inflammatory biomarkers, coagulation parameters, blood counts, and others. Individual meta-analysis was performed for every evaluation endpoint:7 days, 30 days, 3 months, 6 months, and 1 year. Meta-analyses were performed using Random Effect Mean-Difference with a 95% Confidence Interval for continuous data and visualized as forest plots in RevMan version 5.3 software. Cochrane Tool to Assess Risk of Bias in Cohort Studies was used to assess potential risk of bias of the included studies. GRADE Profiler was used to assess quality of evidence. RESULTS Seventy-seven studies fulfilled the inclusion criteria. Surviving SICH patients have significantly lower C-reactive protein (CRP), D-dimer, copeptin, S100β, white blood cell (WBC), monocyte, and glucose than non-surviving patients. SICH patients with good functional outcome have lower D-dimer, Interleukin 6 (IL-6), tumor necrosis factor α (TNF-α), WBC count, neutrophil count, monocyte count, copeptin and significantly higher lymphocyte counts and calcium levels. Out of all blood biomarkers that were evaluated, only S100β and copeptin had very high effect size and high certainty of evidence. CONCLUSION It is interesting to notice that many blood biomarkers significantly associated with SICH patients' outcomes are related to inflammatory responses. This suggests that modulation of inflammation might be essential to improve SICH patients' prognosis. We confidently concluded that S100β and copeptin are the most reliable blood biomarkers that can be used as prognosticators in SICH patients. On other biomarkers, in addition to heterogeneities and inconsistencies, several factors might affect the conclusions of current meta-analysis; thus, future studies to increase the certainties of evidence and effect size on other biomarkers are crucial.
Collapse
Affiliation(s)
- Aloysius Bagus Sasongko
- Department of Neurosurgery, Faculty of Medicine, Pelita Harapan University / Siloam Hospitals, Tangerang, Banten, Indonesia
- Post Graduate Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
| | - Petra Octavian Perdana Wahjoepramono
- Department of Neurosurgery, Faculty of Medicine, Pelita Harapan University / Siloam Hospitals, Tangerang, Banten, Indonesia
- Post Graduate Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
| | - Danny Halim
- Department of Neurosurgery, Faculty of Medicine, Universitas Padjadjaran / Dr. Hasan Sadikin General Hospital, Bandung, West Java, Indonesia
- Research Centre for Medical Genetics, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
| | - Jenifer Kiem Aviani
- Research Centre for Medical Genetics, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
| | - Achmad Adam
- Department of Neurosurgery, Faculty of Medicine, Universitas Padjadjaran / Dr. Hasan Sadikin General Hospital, Bandung, West Java, Indonesia
| | - Yeo Tseng Tsai
- Division of Neurosurgery, Department of Surgery, National University Hospital, Singapore, Singapore
| | - Eka Julianta Wahjoepramono
- Department of Neurosurgery, Faculty of Medicine, Pelita Harapan University / Siloam Hospitals, Tangerang, Banten, Indonesia
| | - Julius July
- Department of Neurosurgery, Faculty of Medicine, Pelita Harapan University / Siloam Hospitals, Tangerang, Banten, Indonesia
| | - Tri Hanggono Achmad
- Research Centre for Medical Genetics, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
- Department of Basic Medical Science, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
| |
Collapse
|
2
|
Mishra S, Grewal J, Wal P, Bhivshet GU, Tripathi AK, Walia V. Therapeutic potential of vasopressin in the treatment of neurological disorders. Peptides 2024; 174:171166. [PMID: 38309582 DOI: 10.1016/j.peptides.2024.171166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/18/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024]
Abstract
Vasopressin (VP) is a nonapeptide made of nine amino acids synthesized by the hypothalamus and released by the pituitary gland. VP acts as a neurohormone, neuropeptide and neuromodulator and plays an important role in the regulation of water balance, osmolarity, blood pressure, body temperature, stress response, emotional challenges, etc. Traditionally VP is known to regulate the osmolarity and tonicity. VP and its receptors are widely expressed in the various region of the brain including cortex, hippocampus, basal forebrain, amygdala, etc. VP has been shown to modulate the behavior, stress response, circadian rhythm, cerebral blood flow, learning and memory, etc. The potential role of VP in the regulation of these neurological functions have suggested the therapeutic importance of VP and its analogues in the management of neurological disorders. Further, different VP analogues have been developed across the world with different pharmacotherapeutic potential. In the present work authors highlighted the therapeutic potential of VP and its analogues in the treatment and management of various neurological disorders.
Collapse
Affiliation(s)
- Shweta Mishra
- SGT College of Pharmacy, SGT University, Gurugram, India
| | - Jyoti Grewal
- Maharisi Markandeshwar University, Sadopur, India
| | - Pranay Wal
- Pranveer Singh Institute of Pharmacy, Kanpur, India
| | | | | | - Vaibhav Walia
- SGT College of Pharmacy, SGT University, Gurugram, India.
| |
Collapse
|
3
|
Sunny A, James RR, Menon SR, Rayaroth S, Daniel A, Thompson NA, Tharakan B. Matrix Metalloproteinase-9 inhibitors as therapeutic drugs for traumatic brain injury. Neurochem Int 2024; 172:105642. [PMID: 38008261 DOI: 10.1016/j.neuint.2023.105642] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/04/2023] [Accepted: 11/09/2023] [Indexed: 11/28/2023]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of morbidity and mortality among young adults and the elderly. In the United States, TBI is responsible for around 30 percent of all injuries brought on by injuries in general. Vasogenic cerebral edema due to blood-brain barrier (BBB) dysfunction and the associated elevation of intracranial pressure (ICP) are some of the major causes of secondary injuries following traumatic brain injury. Matrix metalloproteinase-9 (MMP-9) is a therapeutic target for being an enzyme that degrades the proteins that make up a part of the microvascular basal lamina as well as inter-endothelial tight junctions of the blood-brain barrier. MMP-9-mediated BBB dysfunctions and the compromise of the BBB is a major pathway that leads the development of vasogenic cerebral edema, elevation of ICP, poor cerebral perfusion and brain herniation following traumatic brain injury. That makes MMP-9 an effective therapeutic target and endogenous or exogenous MMP-9 inhibitors as therapeutic drugs for preventing secondary brain damage after traumatic brain injury. Although our understanding of the mechanisms that underlie the primary and secondary stages of damage following a TBI has significantly improved in recent years, such information has not yet resulted in the successful development of novel pharmacological treatment options for traumatic brain injury. Recent pre-clinical and/or clinical studies have demonstrated that there are several compounds with specific or non-specific MMP-9 inhibitory properties either directly binding and inhibiting MMP-9 or by indirectly inhibiting MMP-9, with potential as therapeutic agents for traumatic brain injury. This article reviews the efficacy of several such medications and potential agents that include endogenous and exogeneous compounds that are at various levels of research and development. MMP-9-based therapeutic drug development has enormous potential in the pharmacological treatment of cerebral edema and/or neuronal injury resulting from traumatic brain injury.
Collapse
Affiliation(s)
- Angel Sunny
- Icahn School of Medicine at Mount Sinai, Elmhurst, NY, USA
| | | | | | | | - Abhijith Daniel
- Pushpagiri Institute of Medical Sciences and Research Centre, Thiruvalla, India
| | - Namita Ann Thompson
- Pushpagiri Institute of Medical Sciences and Research Centre, Thiruvalla, India
| | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
4
|
Zhou M, Jiao Q, Wu Z, Li W, Liu G, Wang R, Tang Y. Uncovering the Oxidative Stress Mechanisms and Targets in Alzheimer's Disease by Integrating Phenotypic Screening Data and Polypharmacology Networks. J Alzheimers Dis 2024; 99:S139-S156. [PMID: 36744334 DOI: 10.3233/jad-220727] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background The oxidative stress hypothesis is challenging the dominant position of amyloid-β (Aβ) in the field of understanding the mechanisms of Alzheimer's disease (AD), a complicated and untreatable neurodegenerative disease. Objective The goal of the present study was to uncover the oxidative stress mechanisms causing AD, as well as the potential therapeutic targets and neuroprotective drugs against oxidative stress mechanisms. Methods In this study, a systematic workflow combining pharmacological experiments and computational prediction was proposed. 222 drugs and natural products were collected first and then tested on SH-SY5Y cells to obtain phenotypic screening data on neuroprotection. The preliminary screening data were integrated with drug-target interactions (DTIs) and multi-scale biomedical data, which were analyzed with statistical tests and gene set enrichment analysis. A polypharmacology network was further constructed for investigation. Results 340 DTIs were matched in multiple databases, and 222 cell viability ratios were calculated for experimental compounds. We identified significant potential therapeutic targets based on oxidative stress mechanisms for AD, including NR3C1, SHBG, ESR1, PGR, and AVPR1A, which might be closely related to neuroprotective effects and pathogenesis. 50% of the top 14 enriched pathways were found to correlate with AD, such as arachidonic acid metabolism and neuroactive ligand-receptor interaction. Several approved drugs in this research were also found to exert neuroprotective effects against oxidative stress mechanisms, including beclometasone, methylprednisolone, and conivaptan. Conclusion Our results indicated that NR3C1, SHBG, ESR1, PGR, and AVPR1A were promising therapeutic targets and several drugs may be repurposed from the perspective of oxidative stress and AD.
Collapse
Affiliation(s)
- Moran Zhou
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Qian Jiao
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Zengrui Wu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Weihua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Guixia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Rui Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
5
|
Dutt M, Kumar A, Rout M, Dehury B, Martinez G, Ndishimye P, Kelvin AA, Kelvin DJ. Drug repurposing for Mpox: Discovery of small molecules as potential inhibitors against DNA-dependent RNA polymerase using molecular modeling approach. J Cell Biochem 2023; 124:701-715. [PMID: 36946432 PMCID: PMC10473176 DOI: 10.1002/jcb.30397] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/23/2023]
Abstract
Mpox (formerly Monkeypox), a zoonotic illness caused by the Mpox virus, belongs to the Orthopoxvirus genus in the family Poxviridae. To design and develop effective antiviral therapeutics against DNA viruses, the DNA-dependent RNA polymerase (DdRp) of poxviruses has emerged as a promising drug target. In the present study, we modeled the three-dimensional (3D) structure of DdRp using a template-based homology approach. After modeling, virtual screening was performed to probe the molecular interactions between 1755 Food and Drug Administration-approved small molecule drugs (≤500 molecular weight) and the DdRp of Mpox. Based on the binding affinity and molecular interaction patterns, five drugs, lumacaftor (-11.7 kcal/mol), conivaptan (-11.7 kcal/mol), betulinic acid (-11.6 kcal/mol), fluspirilene (-11.3 kcal/mol), and imatinib (-11.2 kcal/mol), have been ranked as the top drug compounds interacting with Mpox DdRp. Complexes of these shortlisted drugs with DdRp were further evaluated using state-of-the-art all-atoms molecular dynamics (MD) simulations on 200 nanoseconds followed by principal component analysis (PCA). MD simulations and PCA results revealed highly stable interactions of these small drugs with DdRp. After due validation in wet-lab using available in vitro and in vivo experiments, these repurposed drugs can be further utilized for the treatment of contagious Mpox virus. The outcome of this study may establish a solid foundation to screen repurposed and natural compounds as potential antiviral therapeutics against different highly pathogenic viruses.
Collapse
Affiliation(s)
- Mansi Dutt
- Department of Microbiology and Immunology, Department of Paediatrics, IWK Health Center, Canadian Centre for Vaccinology (CCfV), Faculty of MedicineDalhousie UniversityHalifaxCanada
| | - Anuj Kumar
- Department of Microbiology and Immunology, Department of Paediatrics, IWK Health Center, Canadian Centre for Vaccinology (CCfV), Faculty of MedicineDalhousie UniversityHalifaxCanada
- European Virus Bioinformatics CenterJenaGermany
| | - Madhusmita Rout
- Bioinformatics DivisionICMR‐Regional Medical Research CentreBhubaneswarOdishaIndia
| | - Budheswar Dehury
- Bioinformatics DivisionICMR‐Regional Medical Research CentreBhubaneswarOdishaIndia
| | - Gustavo Martinez
- Department of Microbiology and Immunology, Department of Paediatrics, IWK Health Center, Canadian Centre for Vaccinology (CCfV), Faculty of MedicineDalhousie UniversityHalifaxCanada
| | - Pacifique Ndishimye
- Department of Microbiology and Immunology, Department of Paediatrics, IWK Health Center, Canadian Centre for Vaccinology (CCfV), Faculty of MedicineDalhousie UniversityHalifaxCanada
| | - Alyson A. Kelvin
- Vaccine and Infectious Disease Organization (VIDO)University of SaskatchewanSaskatoonSaskatchewanCanada
- Department of Biochemistry, Microbiology, and ImmunologyUniversity of SaskatchewanSaskatoonSaskatchewanCanada
| | - David J. Kelvin
- Department of Microbiology and Immunology, Department of Paediatrics, IWK Health Center, Canadian Centre for Vaccinology (CCfV), Faculty of MedicineDalhousie UniversityHalifaxCanada
| |
Collapse
|
6
|
Calvillo-Robledo A, Ramírez-Farías C, Valdez-Urias F, Huerta-Carreón EP, Quintanar-Stephano A. Arginine vasopressin hormone receptor antagonists in experimental autoimmune encephalomyelitis rodent models: A new approach for human multiple sclerosis treatment. Front Neurosci 2023; 17:1138627. [PMID: 36998727 PMCID: PMC10043225 DOI: 10.3389/fnins.2023.1138627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic demyelinating and neurodegenerative disease that affects the central nervous system. MS is a heterogeneous disorder of multiple factors that are mainly associated with the immune system including the breakdown of the blood-brain and spinal cord barriers induced by T cells, B cells, antigen presenting cells, and immune components such as chemokines and pro-inflammatory cytokines. The incidence of MS has been increasing worldwide recently, and most therapies related to its treatment are associated with the development of several secondary effects, such as headaches, hepatotoxicity, leukopenia, and some types of cancer; therefore, the search for an effective treatment is ongoing. The use of animal models of MS continues to be an important option for extrapolating new treatments. Experimental autoimmune encephalomyelitis (EAE) replicates the several pathophysiological features of MS development and clinical signs, to obtain a potential treatment for MS in humans and improve the disease prognosis. Currently, the exploration of neuro-immune-endocrine interactions represents a highlight of interest in the treatment of immune disorders. The arginine vasopressin hormone (AVP) is involved in the increase in blood−brain barrier permeability, inducing the development and aggressiveness of the disease in the EAE model, whereas its deficiency improves the clinical signs of the disease. Therefore, this present review discussed on the use of conivaptan a blocker of AVP receptors type 1a and type 2 (V1a and V2 AVP) in the modulation of immune response without completely depleting its activity, minimizing the adverse effects associated with the conventional therapies becoming a potential therapeutic target in the treatment of patients with multiple sclerosis.
Collapse
|
7
|
Chojnowski K, Opiełka M, Gozdalski J, Radziwon J, Dańczyszyn A, Aitken AV, Biancardi VC, Winklewski PJ. The Role of Arginine-Vasopressin in Stroke and the Potential Use of Arginine-Vasopressin Type 1 Receptor Antagonists in Stroke Therapy: A Narrative Review. Int J Mol Sci 2023; 24:ijms24032119. [PMID: 36768443 PMCID: PMC9916514 DOI: 10.3390/ijms24032119] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/25/2023] Open
Abstract
Stroke is a life-threatening condition in which accurate diagnoses and timely treatment are critical for successful neurological recovery. The current acute treatment strategies, particularly non-invasive interventions, are limited, thus urging the need for novel therapeutical targets. Arginine vasopressin (AVP) receptor antagonists are emerging as potential targets to treat edema formation and subsequent elevation in intracranial pressure, both significant causes of mortality in acute stroke. Here, we summarize the current knowledge on the mechanisms leading to AVP hyperexcretion in acute stroke and the subsequent secondary neuropathological responses. Furthermore, we discuss the work supporting the predictive value of measuring copeptin, a surrogate marker of AVP in stroke patients, followed by a review of the experimental evidence suggesting AVP receptor antagonists in stroke therapy. As we highlight throughout the narrative, critical gaps in the literature exist and indicate the need for further research to understand better AVP mechanisms in stroke. Likewise, there are advantages and limitations in using copeptin as a prognostic tool, and the translation of findings from experimental animal models to clinical settings has its challenges. Still, monitoring AVP levels and using AVP receptor antagonists as an add-on therapeutic intervention are potential promises in clinical applications to alleviate stroke neurological consequences.
Collapse
Affiliation(s)
- Karol Chojnowski
- Student Scientific Circle of the Department of Adult Neurology, Medical University of Gdansk, 17 Smoluchowskiego Street, 80-214 Gdansk, Poland
| | - Mikołaj Opiełka
- Student Scientific Circle of the Department of Adult Neurology, Medical University of Gdansk, 17 Smoluchowskiego Street, 80-214 Gdansk, Poland
| | - Jacek Gozdalski
- Department of Adult Neurology, Medical University of Gdansk, 17 Smoluchowskiego Street, 80-214 Gdansk, Poland
- Correspondence: (J.G.); (P.J.W.)
| | - Jakub Radziwon
- Student Scientific Circle of the Department of Adult Neurology, Medical University of Gdansk, 17 Smoluchowskiego Street, 80-214 Gdansk, Poland
| | - Aleksandra Dańczyszyn
- Student Scientific Circle of the Department of Adult Neurology, Medical University of Gdansk, 17 Smoluchowskiego Street, 80-214 Gdansk, Poland
| | - Andrew Vieira Aitken
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
- Center for Neurosciences Initiative, Auburn University, Auburn, AL 36849, USA
| | - Vinicia Campana Biancardi
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
- Center for Neurosciences Initiative, Auburn University, Auburn, AL 36849, USA
| | - Paweł Jan Winklewski
- Department of Human Physiology, Medical University of Gdansk, 15 Tuwima Street, 80-210 Gdansk, Poland
- 2nd Department of Radiology, Medical University of Gdansk, 17 Smoluchowskiego Street, 80-214 Gdansk, Poland
- Correspondence: (J.G.); (P.J.W.)
| |
Collapse
|
8
|
Wan Y, Holste KG, Hua Y, Keep RF, Xi G. Brain edema formation and therapy after intracerebral hemorrhage. Neurobiol Dis 2023; 176:105948. [PMID: 36481437 PMCID: PMC10013956 DOI: 10.1016/j.nbd.2022.105948] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/28/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
Intracerebral hemorrhage (ICH) accounts for about 10% of all strokes in the United States of America causing a high degree of disability and mortality. There is initial (primary) brain injury due to the mechanical disruption caused by the hematoma. There is then secondary injury, triggered by the initial injury but also the release of various clot-derived factors (e.g., thrombin and hemoglobin). ICH alters brain fluid homeostasis. Apart from the initial hematoma mass, ICH causes blood-brain barrier disruption and parenchymal cell swelling, which result in brain edema and intracranial hypertension affecting patient prognosis. Reducing brain edema is a critical part of post-ICH care. However, there are limited effective treatment methods for reducing perihematomal cerebral edema and intracranial pressure in ICH. This review discusses the mechanisms underlying perihematomal brain edema formation, the effects of sex and age, as well as how edema is resolved. It examines progress in pharmacotherapy, particularly focusing on drugs which have been or are currently being investigated in clinical trials.
Collapse
Affiliation(s)
- Yingfeng Wan
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
9
|
Barkas F, Anastasiou G, Liamis G, Milionis H. A step-by-step guide for the diagnosis and management of hyponatraemia in patients with stroke. Ther Adv Endocrinol Metab 2023; 14:20420188231163806. [PMID: 37033701 PMCID: PMC10074625 DOI: 10.1177/20420188231163806] [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] [Received: 10/02/2022] [Accepted: 02/27/2023] [Indexed: 04/07/2023] Open
Abstract
Hyponatraemia is common in patients with stroke and associated with adverse outcomes and increased mortality risk. The present review presents the underlying causes and provides a thorough algorithm for the diagnosis and management of hyponatraemia in stroke patients. Concomitant diseases and therapies, such as diabetes, chronic kidney disease and heart failure, along with diuretics, antidepressants and proton pump inhibitors are the most common causes of hyponatraemia in community. In the setting of acute stroke, the emergence of hyponatraemia might be attributed to the administration of hypotonic solutions and drugs (ie. mannitol and antiepileptics), poor solute intake, infections, as well as stroke-related conditions or complications, such as the syndrome of inappropriate secretion of antidiuretic hormone, cerebral salt wasting syndrome and secondary adrenal insufficiency. Diagnostically, the initial step is to differentiate hypotonic from non-hypotonic hyponatraemia, usually caused by hyperglycaemia or recent mannitol administration in patients with stroke. Determining urine osmolality, urine sodium level and volume status are the following steps in the differentiation of hypotonic hyponatraemia. Of note, specific parameters, such as fractional uric acid and urea excretion, along with plasma copeptin concentration, may further improve the diagnostic yield. Therapeutic options are based on the duration and symptoms of hyponatremia. In the case of acute or symptomatic hyponatraemia, hypertonic saline administration is recommended. Hypovolaemic chronic hyponatremia is treated with isotonic solution administration. Although fluid restriction remains the first-line treatment for the rest forms of chronic hyponatraemia, therapies increasing renal free water excretion may be necessary. Loop diuretics and urea serve this purpose in patients with stroke, whereas sodium-glucose transport protein-2 inhibitors appear to be a promising therapy. Nevertheless, it is yet unclear whether the appropriate restoration of sodium level improves outcomes in such patients. Randomized trials designed to compare therapeutic strategies in managing hyponatraemia in patients with stroke are required.
Collapse
Affiliation(s)
- Fotios Barkas
- Department of Hygiene and Epidemiology, Faculty of
Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Georgia Anastasiou
- Department of Internal Medicine, Faculty of Medicine,
School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - George Liamis
- Department of Internal Medicine, Faculty of Medicine,
School of Health Sciences, University of Ioannina, Ioannina, Greece
| | | |
Collapse
|
10
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
11
|
Aleksandrowicz M, Kozniewska E. Hyponatremia as a risk factor for microvascular spasm following subarachnoid hemorrhage. Exp Neurol 2022; 355:114126. [DOI: 10.1016/j.expneurol.2022.114126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 05/20/2022] [Accepted: 05/25/2022] [Indexed: 11/04/2022]
|
12
|
Yuen KCJ, Sharf V, Smith E, Kim M, Yuen ASM, MacDonald PR. Sodium and water perturbations in patients who had an acute stroke: clinical relevance and management strategies for the neurologist. Stroke Vasc Neurol 2021; 7:258-266. [PMID: 34969834 PMCID: PMC9240457 DOI: 10.1136/svn-2021-001230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 11/26/2021] [Indexed: 12/17/2022] Open
Abstract
Sodium and water perturbations, manifesting as hyponatraemia and hypernatraemia, are common in patients who had an acute stroke, and are associated with worse outcomes and increased mortality. Other non-stroke-related causes of sodium and water perturbations in these patients include underlying comorbidities and concomitant medications. Additionally, hospitalised patients who had an acute stroke may receive excessive intravenous hypotonic solutions, have poor fluid intake due to impaired neurocognition and consciousness, may develop sepsis or are administered drugs (eg, mannitol); factors that can further alter serum sodium levels. Sodium and water perturbations can also be exacerbated by the development of endocrine consequences after an acute stroke, including secondary adrenal insufficiency, syndrome of inappropriate antidiuretic hormone secretion and diabetes insipidus. Recently, COVID-19 infection has been reported to increase the risk of development of sodium and water perturbations that may further worsen the outcomes of patients who had an acute stroke. Because there are currently no accepted consensus guidelines on the management of sodium and water perturbations in patients who had an acute stroke, we conducted a systematic review of the literature published in English and in peer-reviewed journals between January 2000 and December 2020, according to PRISMA guidelines, to assess on the current knowledge and clinical practices of this condition. In this review, we discuss the signs and symptoms of hyponatraemia and hypernatraemia, the pathogenesis of hyponatraemia and hypernatraemia, their clinical relevance, and we provide our recommendations for effective treatment strategies for the neurologist in the management of sodium and water perturbations in commonly encountered aetiologies of patients who had an acute stroke.
Collapse
Affiliation(s)
- Kevin C J Yuen
- Departments of Neuroendocrinology and Neurosurgery, Barrow Neurological Institute, Phoenix, Arizona, USA .,University of Arizona College of Medicine and Creighton School of Medicine, Phoenix, Arizona, USA
| | - Valerie Sharf
- University of Arizona College of Medicine and Creighton School of Medicine, Phoenix, Arizona, USA.,Department of Neurology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | | | - Minhwan Kim
- Creighton University School of Medicine, Phoenix, Arizona, USA
| | | | - Paul R MacDonald
- University of Arizona College of Medicine and Creighton School of Medicine, Phoenix, Arizona, USA.,Department of Neurology, Barrow Neurological Institute, Phoenix, Arizona, USA
| |
Collapse
|
13
|
Li D, Cui D, Jia S, Liu X, Wang X, Qiu D, Wang YF. Involvement of Supraoptic Astrocytes in Basilar Artery Occlusion-Evoked Differential Activation of Vasopressin Neurons and Vasopressin Secretion in Rats. Neurochem Res 2021; 46:2651-2661. [PMID: 33532897 DOI: 10.1007/s11064-021-03246-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/02/2021] [Accepted: 01/12/2021] [Indexed: 10/22/2022]
Abstract
Vasopressin (VP) is a key factor in the development of brain injury in ischemic stroke. However, the regulation of VP secretion in basilar artery occlusion (BAO) remains unclear. To clarify the regulation of VP secretion in BAO and the underlying mechanisms, we performed this study in a rat model of BAO with (BC) or without common carotid artery occlusion (CCAO). The results showed that BAO and BC time-dependently increased neurological scores and that BC also increased water contents in the medulla at 2 h and in the pontine at 8 h. Moreover, plasma VP level increased significantly at BAO-8 h, CCAO and BC-2 h but not at BC-8 h; however, VP expressions increased in the supraoptic nucleus (SON) at BC-8 h. The neurological scores were highly correlated with pontine water contents and plasma VP levels. The number of phosphorylated extracellular signal-regulated protein kinase1/2-positive VP neurons increased significantly in the SON at BC-8 h. Similarly, the number of c-Fos-positive VP neurons increased significantly in the SON at BAO-8 h and BC-8 h. In addition, the length of glial fibrillary acidic protein (GFAP) filaments increased significantly in BC compared to BAO only. Aquaporin 4 (AQP4) puncta around VP neurons increased significantly at BC-8 h relative to BC-2 h, which had negative correlation with plasma VP levels. These findings indicate that BAO facilitates VP secretion and increases VP neuronal activity in the SON. The peripheral VP release is possibly under a negative feedback regulation of central VP neuronal activity through increasing GFAP and AQP4 expression in astrocytic processes.
Collapse
Affiliation(s)
- Dongyang Li
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, 157 Baojian Road, Nangang, Harbin, 150081, China.
- Department of Physiology, Hainan Medical University, Haikou, China.
| | - Dan Cui
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, 157 Baojian Road, Nangang, Harbin, 150081, China
| | - Shuwei Jia
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, 157 Baojian Road, Nangang, Harbin, 150081, China
| | - Xiaoyu Liu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, 157 Baojian Road, Nangang, Harbin, 150081, China
| | - Xiaoran Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, 157 Baojian Road, Nangang, Harbin, 150081, China
| | - Delai Qiu
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Yu-Feng Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, 157 Baojian Road, Nangang, Harbin, 150081, China.
| |
Collapse
|
14
|
Cui D, Jia S, Yu J, Li D, Li T, Liu Y, Chang J, Wang X, Liu X, Wang YF. Alleviation of Cerebral Infarction of Rats With Middle Cerebral Artery Occlusion by Inhibition of Aquaporin 4 in the Supraoptic Nucleus. ASN Neuro 2021; 12:1759091420960550. [PMID: 32985231 PMCID: PMC7545515 DOI: 10.1177/1759091420960550] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
In ischemic stroke, vasopressin hypersecretion is a critical factor of cerebral swelling and brain injury. To clarify neural mechanisms underlying ischemic stroke-evoked vasopressin hypersecretion, we observed the effect of unilateral permanent middle cerebral artery occlusion (MCAO) in rats on astrocytic plasticity and vasopressin neuronal activity in the supraoptic nucleus (SON) as well as their associated cerebral injuries. MCAO for 8 hr caused cerebral infarction in the MCAO side where water contents also increased. Immunohistochemical examination revealed that the percentage of phosphorylated extracellular signal-regulated protein kinase 1/2 (pERK1/2)-positive vasopressin neurons in the SON of MCAO side was significantly higher than that in non-MCAO side and in sham group. In the cortex, pERK1/2 and aquaporin 4 expressions increased significantly in the infarction area, while glial fibrillary acidic protein (GFAP) reduced significantly compared with the noninfarction side in brain cortex. Microinjection of N-(1,3,4-Thiadiazolyl)nicotinamide-020 [TGN-020, a specific blocker of aquaporin 4] into the SON blocked MCAO-evoked increases in pERK1/2 in the SON as well as the reduction of GFAP and the increase in pERK1/2 and aquaporin 4 in the infarction area of the cortex. Finally, oxygen and glucose deprivation reduced GFAP expression and the colocalization and molecular association of GFAP with aquaporin 4 in the SON in brain slices. These effects were blocked by TGN-020 and/or phloretin, a blocker of astrocytic volume-regulated anion channels. These findings indicate that blocking aquaporin 4 in the SON may reduce the activation of vasopressin neurons and brain injuries elicited by vasopressin during ischemic stroke.
Collapse
Affiliation(s)
- Dan Cui
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Shuwei Jia
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Jiawei Yu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Dongyang Li
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Tong Li
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Yang Liu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Jinlong Chang
- The Seventh Affiliated Hospital, Sun Yat-sen Universtiy, Shenzhen, China
| | - Xiaoran Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Xiaoyu Liu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Yu-Feng Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| |
Collapse
|
15
|
Stokum JA, Gerzanich V, Sheth KN, Kimberly WT, Simard JM. Emerging Pharmacological Treatments for Cerebral Edema: Evidence from Clinical Studies. Annu Rev Pharmacol Toxicol 2020; 60:291-309. [PMID: 31914899 DOI: 10.1146/annurev-pharmtox-010919-023429] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cerebral edema, a common and often fatal companion to most forms of acute central nervous system disease, has been recognized since the time of ancient Egypt. Unfortunately, our therapeutic armamentarium remains limited, in part due to historic limitations in our understanding of cerebral edema pathophysiology. Recent advancements have led to a number of clinical trials for novel therapeutics that could fundamentally alter the treatment of cerebral edema. In this review, we discuss these agents, their targets, and the data supporting their use, with a focus on agents that have progressed to clinical trials.
Collapse
Affiliation(s)
- Jesse A Stokum
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA;
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA;
| | - Kevin N Sheth
- Department of Neurology, Division of Neurocritical Care and Emergency Neurology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - W Taylor Kimberly
- Department of Neurology, Division of Neurocritical Care, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA; .,Departments of Pathology and Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| |
Collapse
|
16
|
Bicker J, Alves G, Fonseca C, Falcão A, Fortuna A. Repairing blood-CNS barriers: Future therapeutic approaches for neuropsychiatric disorders. Pharmacol Res 2020; 162:105226. [PMID: 33007420 DOI: 10.1016/j.phrs.2020.105226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 12/19/2022]
Abstract
Central nervous system (CNS) drug development faces significant difficulties that translate into high rates of failure and lack of innovation. The pathophysiology of neurological and psychiatric disorders often results in the breakdown of blood-CNS barriers, disturbing the CNS microenvironment and worsening disease progression. Therefore, restoring the integrity of blood-CNS barriers may have a beneficial influence in several CNS disorders and improve treatment outcomes. In this review, pathways that may be modulated to protect blood-CNS barriers from neuroinflammatory and oxidative insults are featured. First, the participation of the brain endothelium and glial cells in disruption processes is discussed. Then, the relevance of regulatory systems is analysed, specifically the hypothalamic-pituitary axis, the renin-angiotensin system, sleep and circadian rhythms, and glutamate neurotransmission. Lastly, compounds of endogenous and exogenous origin that are known to mediate the repair of blood-CNS barriers are presented. We believe that enhancing the protection of blood-CNS barriers is a promising therapeutic strategy to pursue in the future.
Collapse
Affiliation(s)
- Joana Bicker
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal.
| | - Gilberto Alves
- CICS-UBI, Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | - Carla Fonseca
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
| | - Amílcar Falcão
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Ana Fortuna
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| |
Collapse
|
17
|
Blockade of Arginine Vasopressin receptors prevents blood-brain barrier breakdown in Experimental Autoimmune Encephalomyelitis. Sci Rep 2020; 10:467. [PMID: 31949182 PMCID: PMC6965180 DOI: 10.1038/s41598-019-57134-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/16/2019] [Indexed: 01/10/2023] Open
Abstract
The blood-brain barrier (BBB) plays a significant pathophysiological role in multiple sclerosis (MS). Vasopressin (AVP) is released after brain injury and contributes to the inflammatory response. We propose that AVP may be modulating BBB permeability and hence affecting EAE clinical signs. Female Lewis rats were immunized s.c. with guinea-pig brain extract suspended in complete Freund’s adjuvant. Prior to that, animals were subjected to Neurointermediate pituitary lobectomy (NIL) or treated with AVP receptor antagonist (conivaptan). BBB permeability assays were performed. Western blot for claudin-5 and histological analysis were performed in conivaptan treated EAE rats. EAE increase in BBB permeability to Evans blue was reverted by the NIL surgery. AVP receptor blockade reverted the EAE BBB hyperpermeability to Evans blue and 10-kDa FITC-dextran in almost all brain regions. Both, AVP low levels and AVP receptor blockade attenuated EAE clinical signs. Conivaptan reduced perivascular cuffs in EAE rats. A decrease in claudin-5 expression was observed in EAE rats and conivaptan treatment partially restored normal levels. Our data indicate that V1a and V2 AVP receptors can modulate BBB permeability and consequently are involved in the CNS inflammatory process during EAE. Future research is required to characterize the utility of vasopressin antagonist in MS treatment.
Collapse
|
18
|
Zeynalov E, Jones SM, Elliott JP. Vasopressin and vasopressin receptors in brain edema. VITAMINS AND HORMONES 2020; 113:291-312. [DOI: 10.1016/bs.vh.2019.08.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
19
|
Spagnolello O, De Michele M, Lorenzano S, Cerulli Irelli E, Naitana F, Falcou A, Letteri F, Bachetoni A, Collepardo D, Bertazzoni G, Toni D. Copeptin Kinetics in Acute Ischemic Stroke May Differ According to Revascularization Strategies: Pilot Data. Stroke 2019; 50:3632-3635. [PMID: 31630623 DOI: 10.1161/strokeaha.119.025433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose- Prognostic value of copeptin in acute ischemic stroke has been widely reported. This study aimed to evaluate copeptin temporal profile according to revascularization strategies and the development of brain edema and hemorrhagic transformation. Methods- Plasma copeptin and brain edema and hemorrhagic transformation assessed by computed tomography/magnetic resonance imaging were evaluated upon admission (T0), at 24 hours (T1), and between the third and fifth day of hospitalization (T2) in 34 acute ischemic stroke patients. Results- Median copeptin concentration was 50.71 pmol/L at T0, 18.31 pmol/L at T1, and 10.92 pmol/L at T2. Copeptin at T1 was higher in patients with medium/severe brain edema at T2 (32.25 versus 13.67 pmol/L; P=0.038) and hemorrhagic transformation at T1 (93.10 versus 13.67 pmol/L; P<0.003) and T2 (85.70 versus 14.45 pmol/L; P=0.024). Copeptin level drop (CopΔT1-T0) was significantly steeper in patients receiving revascularization, particularly in those undergoing combined therapy (-129.34 versus -5.43 pmol/L; P=0.038). ΔT1-T0 also correlated with Thrombolysis in Cerebral Infarction score (P<0.001). Conclusions- Copeptin resulted associated with brain edema and hemorrhagic transformation in acute ischemic stroke, and its drop at 24 hours may mirror effective brain vessel recanalization.
Collapse
Affiliation(s)
- Ornella Spagnolello
- From the Emergency Department (O.S., F.N., G.B.), Policlinico Umberto I Hospital, Sapienza University, Rome, Italy
| | - Manuela De Michele
- Department of Human Neurosciences (M.D.M., S.L., E.C.I., A.F., F.L., D.T.), Policlinico Umberto I Hospital, Sapienza University, Rome, Italy
| | - Svetlana Lorenzano
- Department of Human Neurosciences (M.D.M., S.L., E.C.I., A.F., F.L., D.T.), Policlinico Umberto I Hospital, Sapienza University, Rome, Italy
| | - Emanuele Cerulli Irelli
- Department of Human Neurosciences (M.D.M., S.L., E.C.I., A.F., F.L., D.T.), Policlinico Umberto I Hospital, Sapienza University, Rome, Italy
| | - Federico Naitana
- From the Emergency Department (O.S., F.N., G.B.), Policlinico Umberto I Hospital, Sapienza University, Rome, Italy
| | - Anne Falcou
- Department of Human Neurosciences (M.D.M., S.L., E.C.I., A.F., F.L., D.T.), Policlinico Umberto I Hospital, Sapienza University, Rome, Italy
| | - Federica Letteri
- Department of Human Neurosciences (M.D.M., S.L., E.C.I., A.F., F.L., D.T.), Policlinico Umberto I Hospital, Sapienza University, Rome, Italy
| | - Alessandra Bachetoni
- Department of Experimental Medicine (A.B., D.C.), Policlinico Umberto I Hospital, Sapienza University, Rome, Italy
| | - Daniela Collepardo
- Department of Experimental Medicine (A.B., D.C.), Policlinico Umberto I Hospital, Sapienza University, Rome, Italy
| | - Giuliano Bertazzoni
- From the Emergency Department (O.S., F.N., G.B.), Policlinico Umberto I Hospital, Sapienza University, Rome, Italy
| | - Danilo Toni
- Department of Human Neurosciences (M.D.M., S.L., E.C.I., A.F., F.L., D.T.), Policlinico Umberto I Hospital, Sapienza University, Rome, Italy
| |
Collapse
|
20
|
Alrabiah H, Bakheit A, Attia S, Mostafa GA. Bioanalytical Method Development and Validation for the Determination of Vasopressin Receptor Antagonist Conivaptan in Mouse Plasma at NanoLevel and its Pharmacokinetic Application. CURR ANAL CHEM 2019. [DOI: 10.2174/1573411014666180330160158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Conivaptan inhibits two of vasopressin receptor (vasopressin receptor V1a and
V2). Conivaptan is used for the treatment of hyponatremia, and in some instances, for the treatment of
the heart failure.
Methods:
The present study aimed to develop a simple, sensitive, and accurate HPLC with ultraviolet
detection for the assay of conivaptan (CON) in mouse plasma using bisoprolol as internal standard (IS).
A precipitation procedure was used to extract CON and the IS from the mouse plasma. CON was chromatographically
separated using a C18 analytical column at 25°C. The separation was carried out using
a mixture of phosphate buffer (50 mM): acetonitrile (60: 40, v/v, pH 4.5) with a flow rate of 1.0 mL/min
and detection was performed at 240 nm.
Results:
The assay was validated according to the US Food and Drug (FDA) guidelines. The method
demonstrated linearity over a concentration range of 150 - 2000 ng/mL (correlation coefficient: r 2 =
0.9985). The mean recovery of CON from the mouse plasma was 101.13%. All validation parameters
for CON were within the acceptable range.
Conclusion:
The investigated method has been shown to be suitable for estimating the CON in plasma
samples, and this method is sensitive and highly selective, allowing the estimation of its concentrations
up to the nano-scale. The suggested method was successfully used in a pharmacokinetic study of CON
in mouse plasma.
Collapse
Affiliation(s)
- Haitham Alrabiah
- Pharmaceutical Chemistry Department, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Ahmed Bakheit
- Pharmaceutical Chemistry Department, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Sabray Attia
- Department of Pharmacology Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457 Riyadh, 11451, Saudi Arabia
| | - Gamal A.E. Mostafa
- Pharmaceutical Chemistry Department, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
21
|
Li W, Li R, Zhao S, Jiang C, Liu Z, Tang X. Lithium Posttreatment Alleviates Blood–Brain Barrier Injury After Intracerebral Hemorrhage in Rats. Neuroscience 2018; 383:129-137. [DOI: 10.1016/j.neuroscience.2018.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/02/2018] [Accepted: 05/01/2018] [Indexed: 10/16/2022]
|
22
|
Wang YF, Parpura V. Astroglial Modulation of Hydromineral Balance and Cerebral Edema. Front Mol Neurosci 2018; 11:204. [PMID: 29946238 PMCID: PMC6007284 DOI: 10.3389/fnmol.2018.00204] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 05/22/2018] [Indexed: 12/11/2022] Open
Abstract
Maintenance of hydromineral balance (HB) is an essential condition for life activity at cellular, tissue, organ and system levels. This activity has been considered as a function of the osmotic regulatory system that focuses on hypothalamic vasopressin (VP) neurons, which can reflexively release VP into the brain and blood to meet the demand of HB. Recently, astrocytes have emerged as an essential component of the osmotic regulatory system in addition to functioning as a regulator of the HB at cellular and tissue levels. Astrocytes express all the components of osmoreceptors, including aquaporins, molecules of the extracellular matrix, integrins and transient receptor potential channels, with an operational dynamic range allowing them to detect and respond to osmotic changes, perhaps more efficiently than neurons. The resultant responses, i.e., astroglial morphological and functional plasticity in the supraoptic and paraventricular nuclei, can be conveyed, physically and chemically, to adjacent VP neurons, thereby influencing HB at the system level. In addition, astrocytes, particularly those in the circumventricular organs, are involved not only in VP-mediated osmotic regulation, but also in regulation of other osmolality-modulating hormones, including natriuretic peptides and angiotensin. Thus, astrocytes play a role in local/brain and systemic HB. The adaptive astrocytic reactions to osmotic challenges are associated with signaling events related to the expression of glial fibrillary acidic protein and aquaporin 4 to promote cell survival and repair. However, prolonged osmotic stress can initiate inflammatory and apoptotic signaling processes, leading to glial dysfunction and a variety of brain diseases. Among many diseases of brain injury and hydromineral disorders, cytotoxic and osmotic cerebral edemas are the most common pathological manifestation. Hyponatremia is the most common cause of osmotic cerebral edema. Overly fast correction of hyponatremia could lead to central pontine myelinolysis. Ischemic stroke exemplifies cytotoxic cerebral edema. In this review, we summarize and analyze the osmosensory functions of astrocytes and their implications in cerebral edema.
Collapse
Affiliation(s)
- Yu-Feng Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
23
|
Zhang J, Li J, Ma L, Lou J. Retracted
: RNA interference‐mediated silencing of S100B improves nerve function recovery and inhibits hippocampal cell apoptosis in rat models of ischemic stroke. J Cell Biochem 2018; 119:8095-8111. [DOI: 10.1002/jcb.26747] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 01/25/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Jin‐Hua Zhang
- Department of NeurologyThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhouP.R. China
- Department of NeurologyKaifeng Central HospitalKaifengP.R. China
| | - Jiang‐Kun Li
- Department of NeurologyKaifeng Central HospitalKaifengP.R. China
| | - Li‐Li Ma
- Department of NeurologyKaifeng Central HospitalKaifengP.R. China
| | - Ji‐Yu Lou
- Department of NeurologyThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhouP.R. China
| |
Collapse
|
24
|
Hayman EG, Wessell A, Gerzanich V, Sheth KN, Simard JM. Mechanisms of Global Cerebral Edema Formation in Aneurysmal Subarachnoid Hemorrhage. Neurocrit Care 2017; 26:301-310. [PMID: 27995510 DOI: 10.1007/s12028-016-0354-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
A growing body of clinical literature emphasizes the impact of cerebral edema in early brain injury following aneurysmal subarachnoid hemorrhage (aSAH). Aneurysm rupture itself initiates global cerebral edema in up to two thirds of cases. Although cerebral edema is not a universal feature of aSAH, it portends a poor clinical course, with quantitative analysis revealing a direct correlation between cerebral edema and poor outcome, including mortality and cognitive deficits. Mechanistically, global cerebral edema has been linked to global ischemia at the time of aneurysm rupture, dysfunction of autoregulation, blood breakdown products, neuroinflammation, and hyponatremia/endocrine abnormalities. At a molecular level, several culprits have been identified, including aquaporin-4, matrix metalloproteinase-9, SUR1-TRPM4 cation channels, vascular endothelial growth factor, bradykinin, and others. Here, we review these cellular and molecular mechanisms of global cerebral edema formation in aSAH. Given the importance of edema to the outcome of patients with aSAH and its status as a highly modifiable pathological process, a better understanding of cerebral edema in aSAH promises to hasten the development of medical therapies to improve outcomes in this frequently devastating disease.
Collapse
Affiliation(s)
- Erik G Hayman
- Department of Neurosurgery, University of Maryland School of Medicine, 22 S. Greene St., Suite S12D, Baltimore, MD, 21201-1595, USA
| | - Aaron Wessell
- Department of Neurosurgery, University of Maryland School of Medicine, 22 S. Greene St., Suite S12D, Baltimore, MD, 21201-1595, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, 22 S. Greene St., Suite S12D, Baltimore, MD, 21201-1595, USA
| | - Kevin N Sheth
- Department of Neurology, Yale New Haven Hospital, New Haven, CT, USA.,Department of Neurosurgery, Yale New Haven Hospital, New Haven, CT, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, 22 S. Greene St., Suite S12D, Baltimore, MD, 21201-1595, USA. .,Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA. .,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
25
|
Zeynalov E, Jones SM, Elliott JP. Therapeutic time window for conivaptan treatment against stroke-evoked brain edema and blood-brain barrier disruption in mice. PLoS One 2017; 12:e0183985. [PMID: 28854286 PMCID: PMC5576654 DOI: 10.1371/journal.pone.0183985] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 08/15/2017] [Indexed: 11/18/2022] Open
Abstract
Background Ischemic stroke is often complicated by brain edema, disruption of blood-brain barrier (BBB), and uncontrolled release of arginine-vasopressin (AVP). Conivaptan, a V1a and V2 receptor antagonist, reduces brain edema and minimizes damage to the blood-brain barrier after stroke. Most stroke patients do not receive treatment immediately after the onset of brain ischemia. Delays in therapy initiation may worsen stroke outcomes. Therefore, we designed a translational study to explore the therapeutic time window for conivaptan administration. Methods Mice were treated with conivaptan beginning 3, 5, or 20 hours after 60-minute focal middle cerebral artery occlusion. Treatments were administered by continuous IV infusion for a total of 48 hours. Brain edema and blood-brain barrier (BBB) disruption were evaluated at endpoint. Results Conivaptan therapy initiated at 3 hours following ischemia reduced edema in the ipsilateral hemisphere, which corresponded with improvements in neurological deficits. Stroke-triggered BBB disruption was also reduced in mice when conivaptan treatments were initiated at 3 hours of reperfusion. However, 5 and 20-hour delays of conivaptan administration failed to reduce edema or protect BBB. Conclusion Timing of conivaptan administration is important for successful reduction of brain edema and BBB disruption. Our experimental data open new possibilities to repurpose conivaptan, and make an important “bench-to-bedside translation” of the results into clinical practice.
Collapse
Affiliation(s)
- Emil Zeynalov
- Swedish Medical Center, Neurotrauma Research, Englewood, Colorado, United States of America
- * E-mail:
| | - Susan M. Jones
- Swedish Medical Center, Neurotrauma Research, Englewood, Colorado, United States of America
| | - J. Paul Elliott
- Colorado Brain and Spine Institute, Englewood, Colorado, United States of America
| |
Collapse
|
26
|
Ji Y, Teng L, Zhang R, Sun J, Guo Y. NRG-1β exerts neuroprotective effects against ischemia reperfusion-induced injury in rats through the JNK signaling pathway. Neuroscience 2017; 362:13-24. [PMID: 28843994 DOI: 10.1016/j.neuroscience.2017.08.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 08/15/2017] [Accepted: 08/15/2017] [Indexed: 01/30/2023]
Abstract
BACKGROUND Neuregulin-1β (NRG-1β) has great potential to be developed into therapeutics for neuroprotection. The aim of the current study was to analyze the effects and possible signaling pathway of NRG-1β on brain tissues in a rat model of middle cerebral artery occlusion/reperfusion (MCAO/R). METHODS In order to observe the protective effect of NRG-1β on MCAO/R, the neurological deficit and infarct volume were measured using a modified neurological severity score (mNSS) test and by triphenyl tetrazolium chloride (TTC) staining. In order to detect the antagonistic effect of NRG-1β on nerve cells and the blood-brain barrier (BBB), the morphology and structure of cortical brain tissues were observed by Evans Blue (EB) staining, hematoxylin-eosin (H&E) and Nissl staining, in situ cell death detection kit, and transmission electron microscopy (TEM). In order to investigate whether NRG-1β exhibited a significant neuroprotective effect via the JNK signaling pathway, the activity of JNK and the levels of phospho-MKK4, phospho-JNK, pan-JNK and phospho-c-Jun were tested using a JNK activity screening kit, immunofluorescent labeling, and western blot analysis, respectively. RESULTS In the NRG-1β treatment group, accompanied with a decrease in JNK activity, the protein levels of phospho-JNK, phospho-MKK4 and phospho-c-Jun decreased, the ischemia-induced apoptosis decreased, the abnormal morphological structures of nerve cells were ameliorated, the integrity of the BBB was restored, and infarct volume was reduced. At the same time, neurological function was significantly recovered. CONCLUSION NRG-1β exerts a neuroprotective effect through the JNK signaling pathway in MCAO/R rats.
Collapse
Affiliation(s)
- Yaqing Ji
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong, China.
| | - Lei Teng
- Department of Biology, Qingdao University, Qingdao 266021, Shandong, China.
| | - Rui Zhang
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong, China.
| | - Jinping Sun
- Hospital of Qingdao University, Qingdao 266003, Shandong, China.
| | - Yunliang Guo
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong, China.
| |
Collapse
|
27
|
Babadjouni RM, Radwanski RE, Walcott BP, Patel A, Durazo R, Hodis DM, Emanuel BA, Mack WJ. Neuroprotective strategies following intraparenchymal hemorrhage. J Neurointerv Surg 2017; 9:1202-1207. [PMID: 28710084 DOI: 10.1136/neurintsurg-2017-013197] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 06/20/2017] [Accepted: 06/23/2017] [Indexed: 12/23/2022]
Abstract
Intracerebral hemorrhage and, more specifically, intraparenchymal hemorrhage, are devastating disease processes with poor clinical outcomes. Primary injury to the brain results from initial hematoma expansion while secondary hemorrhagic injury occurs from blood-derived products such as hemoglobin, heme, iron, and coagulation factors that overwhelm the brains natural defenses. Novel neuroprotective treatments have emerged that target primary and secondary mechanisms of injury. Nonetheless, translational application of neuroprotectants from preclinical to clinical studies has yet to show beneficial clinical outcomes. This review summarizes therapeutic agents and neuroprotectants in ongoing clinical trials aimed at targeting primary and secondary mechanisms of injury after intraparenchymal hemorrhage.
Collapse
Affiliation(s)
- Robin Moshe Babadjouni
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Ryan E Radwanski
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Brian P Walcott
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Arati Patel
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Ramon Durazo
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Drew M Hodis
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Benjamin A Emanuel
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - William J Mack
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
28
|
Vinod P, Krishnappa V, Chauvin AM, Khare A, Raina R. Cardiorenal Syndrome: Role of Arginine Vasopressin and Vaptans in Heart Failure. Cardiol Res 2017; 8:87-95. [PMID: 28725324 PMCID: PMC5505291 DOI: 10.14740/cr553w] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 05/18/2017] [Indexed: 02/06/2023] Open
Abstract
Heart and kidney failure continued to be of increasing prevalence in today's society, and their comorbidity has synergistic effect on the morbidity and mortality of patients. Cardiorenal syndrome (CRS) is a complex disease with multifactorial pathophysiology. Better understanding of this pathophysiological network is crucial for the successful intervention to prevent advancement of the disease process. One of the major factors in this process is neurohormonal activation, predominantly involving renin-angiotensin-aldosterone system (RAAS) and arginine vasopressin (AVP). Heart failure causes reduced cardiac output/cardiac index (CO/CI) and fall in renal perfusion pressures resulting in activation of baroreceptors and RAAS, respectively. Activated baroreceptors and RAAS stimulate the release of AVP (non-osmotic pathway), which acts on V2 receptors located in the renal collecting ducts, causing fluid retention and deterioration of heart failure. Effective blockade of AVP action on V2 receptors has emerged as a potential treatment option in volume overload conditions especially in the setting of hyponatremia. Vasopressin receptor antagonists (VRAs), such as vaptans, are potent aquaretics causing electrolyte-free water diuresis without significant electrolyte abnormalities. Vaptans are useful in hypervolemic hyponatremic conditions like heart failure and liver cirrhosis, and euvolemic hyponatremic conditions like syndrome of inappropriate anti-diuretic hormone secretion. Tolvaptan and conivaptan are pharmaceutical agents that are available for the treatment of these conditions.
Collapse
Affiliation(s)
- Poornima Vinod
- Department of Internal Medicine, Cleveland Clinic Akron General, Akron, OH, USA
| | - Vinod Krishnappa
- Cleveland Clinic Akron General/Akron Nephrology Associates, Akron, OH, USA
| | | | - Anshika Khare
- Northeast Ohio Medical University, Rootstown, OH, USA
| | - Rupesh Raina
- Department of Nephrology, Cleveland Clinic Akron General, Akron, OH, USA
| |
Collapse
|
29
|
Conivaptan, a Selective Arginine Vasopressin V1a and V2 Receptor Antagonist Attenuates Global Cerebral Edema Following Experimental Cardiac Arrest via Perivascular Pool of Aquaporin-4. Neurocrit Care 2017; 24:273-82. [PMID: 26732270 DOI: 10.1007/s12028-015-0236-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND Cerebral edema is a major cause of mortality following cardiac arrest (CA) and cardiopulmonary resuscitation (CPR). Arginine vasopressin (AVP) and water channel aquaporin-4 (AQP4) have been implicated in the pathogenesis of CA-evoked cerebral edema. In this study, we examined if conivaptan, a V1a and V2 antagonist, attenuates cerebral edema following CA/CPR in wild type (WT) mice as well as mice with targeted disruption of the gene encoding α-syntrophin (α-syn(-/-)) that demonstrate diminished perivascular AQP4 pool. METHODS Isoflurane-anesthetized adult male WT C57Bl/6 and α-syn(-/-) mice were subjected to 8 min CA/CPR and treated with either bolus IV injection (0.15 or 0.3 mg/kg) followed by continuous infusion of conivaptan (0.15 mg/kg/day or 0.3 mg/kg/day), or vehicle infusion for 48 h. Serum osmolality, regional brain water content, and blood-brain barrier (BBB) disruption were determined at the end of the experiment. Sham-operated mice in both strains served as controls. RESULTS Treatment with conivaptan elevated serum osmolality in a dose-dependent manner. In WT mice, conivaptan at 0.3 mg dose significantly attenuated regional water content in the caudoputamen (81.0 ± 0.5 vs. 82.5 ± 0.4% in controls; mean ± SEM) and cortex (78.8 ± 0.2 vs. 79.4 ± 0.2% in controls), while conivaptan at 0.15 mg was not effective. In α-syn(-/-) mice, conivaptan at 0.3 mg dose did not attenuate water content compared with controls. Conivaptan (0.3 mg/kg/day) attenuated post-CA BBB disruption at 48 h in WT mice but not in α-syn(-/-) mice. CONCLUSIONS Continuous IV infusion of conivaptan attenuates cerebral edema and BBB disruption following CA. These effects of conivaptan that are dependent on the presence of perivascular pool of AQP4 appear be mediated via its dual effect on V1 and V2 receptors.
Collapse
|
30
|
da Costa LHA, Júnior NNDS, Catalão CHR, Sharshar T, Chrétien F, da Rocha MJA. Vasopressin Impairment During Sepsis Is Associated with Hypothalamic Intrinsic Apoptotic Pathway and Microglial Activation. Mol Neurobiol 2016; 54:5526-5533. [DOI: 10.1007/s12035-016-0094-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 09/01/2016] [Indexed: 01/09/2023]
|
31
|
Zeynalov E, Jones SM, Elliott JP. Continuous IV Infusion is the Choice Treatment Route for Arginine-vasopressin Receptor Blocker Conivaptan in Mice to Study Stroke-evoked Brain Edema. J Vis Exp 2016. [PMID: 27684044 DOI: 10.3791/54170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Stroke is one of the major causes of morbidity and mortality in the world. Stroke is complicated by brain edema and other pathophysiological events. Among the most important players in the development and evolution of stroke-evoked brain edema is the hormone arginine-vasopressin and its receptors, V1a and V2. Recently, the V1a and V2 receptor blocker conivaptan has been attracting attention as a potential drug to reduce brain edema after stroke. However, animal models which involve conivaptan applications in stroke research need to be modified based on feasible routes of administration. Here the outcomes of 48 hr continuous intravenous (IV) are compared with intraperitoneal (IP) conivaptan treatments after experimental stroke in mice. We developed a protocol in which middle cerebral artery occlusion was combined with catheter installation into the jugular vein for IV treatment of conivaptan (0.2 mg) or vehicle. Different cohorts of animals were treated with 0.2 mg bolus of conivaptan or vehicle IP daily. Experimental stroke-evoked brain edema was evaluated in mice after continuous IV and IP treatments. Comparison of the results revealed that the continuous IV administration of conivaptan alleviates post-ischemic brain edema in mice, unlike the IP administration of conivaptan. We conclude that our model can be used for future studies of conivaptan applications in the context of stroke and brain edema.
Collapse
|
32
|
Wei X, Hu CC, Zhang YL, Yao SL, Mao WK. Telmisartan reduced cerebral edema by inhibiting NLRP3 inflammasome in mice with cold brain injury. ACTA ACUST UNITED AC 2016; 36:576-583. [DOI: 10.1007/s11596-016-1628-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 06/13/2016] [Indexed: 12/11/2022]
|
33
|
Gu J, Chen J, Yang N, Hou X, Wang J, Tan X, Feng L, Jia X. Combination of Ligusticum chuanxiong and Radix Paeoniae ameliorate focal cerebral ischemic in MCAO rats via endoplasmic reticulum stress-dependent apoptotic signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2016; 187:313-324. [PMID: 27108052 DOI: 10.1016/j.jep.2016.04.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/29/2016] [Accepted: 04/19/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Combination of Ligusticum chuanxiong and Radix Paeoniae (XS) is highly effective in the treatment for focal cerebral ischemic, but the underlying mechanism is not clear. This study was conducted to evaluate the combinative effects of XS on MCAO rats and explore the underlying mechanisms. MATERIALS AND METHODS MCAO rats were used to evaluate the protective effect of Ligusticum chuanxiong (CX), Radix Paeoniae Rubra (CS) and their combination (XS) on ameliorating focal cerebral ischemic. Cerebral ischemia deficits and infarct size were performed by 2,3,5-triphenyltetrazolium chloride (TTC) and hematoxylin-eosin (H-E) staining. Activities of SOD, CAT and GSH-Px, as well as levels of LPO and MDA were detected by commercial kits while ELISA kits for the content of plasminogen activator inhibitor-1 (PAI-1) and plasminogen activator (PA). Immunohistochemistry (IHC) and western blot analysis (WB) were carried out to examine the protein expressions including PKR-like endoplasmic reticulum kinase (PERK), cytoplasmic of glucose regulated protein 78 (GRP78), X box-binding protein-1 (XBP-1), activating transcription factor-6 (ATF-6), C/EBP-homologous protein (CHOP), metalloprotease-9 (MMP-9), tissue inhibitor of metalloproteinase-1 (TIMP-1), Bcl-2 associated X protein (Bax), and porcineB-cellleukemia/lymphoma-2 (Bcl-2) in brain tissues. Reverse transcription polymerase chain reaction (RT-PCR) and Quantitative PCR (Q-PCR) were applied to examine vascular endothelial growth factor (VEGF) and N-methyl-d-aspartate receptors (NMDAR1) mRNA levels. RESULTS CX, CS and their combination (XS) could reduce cerebral ischemia deficits and infarct size of MCAO rats. They increased SOD, CAT and GSH-Px activities, and reduced MDA and LPO levels in serum, markedly. A significant decrease of endoplasmic reticulum stress-related factors PERK, XBP-1, ATF-6 and CHOP protein expression levels while an increase of GRP78 and MVD expression by the treatment of CX, CS and XS. It could also be observed that their treatment could reduce apoptotic damage of brain tissues by up-regulating Bax level and down-regulating Bcl-2 level. Furthermore, the levels of MMP-9 and PAI-1 in serum and tissues of rats were down-regulated remarkably while TIMP-1 and PA levels were up-regulated. VEGF mRNA level was up-regulated dramatically whereas NMDAR1 was reduced. Importantly, the combination of CX and CS, namely XS, has a more meaningful improvement on focal cerebral ischemic than CX or CS alone. CONCLUSION All these revealed that the combined XS exerted more remarkable protective effects than alone. XS could inhibit neuronal apoptosis by attenuating ER-stress-dependent apoptotic signaling and protected the blood-brain barrier. These findings might supply beneficial hints for the synergy of CX and CS, and provide the basis for rationality of XS preparation and deserve further clinical investigations.
Collapse
Affiliation(s)
- Junfei Gu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, PR China; State Key Laboratory Breeding Base of Dao-di Herbs, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China; Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, Jiangsu 210028, PR China
| | - Juan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, PR China; Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, Jiangsu 210028, PR China
| | - Nan Yang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, PR China; Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, Jiangsu 210028, PR China
| | - Xuefeng Hou
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, PR China; Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, Jiangsu 210028, PR China
| | - Jing Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, PR China; Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, Jiangsu 210028, PR China
| | - Xiaobin Tan
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, PR China; Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, Jiangsu 210028, PR China
| | - Liang Feng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, PR China; State Key Laboratory Breeding Base of Dao-di Herbs, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China; Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, Jiangsu 210028, PR China.
| | - Xiaobin Jia
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, PR China; Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, Jiangsu 210028, PR China.
| |
Collapse
|
34
|
Zhao JJ, Song JQ, Pan SY, Wang K. Treatment with Isorhamnetin Protects the Brain Against Ischemic Injury in Mice. Neurochem Res 2016; 41:1939-48. [PMID: 27161367 DOI: 10.1007/s11064-016-1904-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 03/22/2016] [Accepted: 03/28/2016] [Indexed: 12/20/2022]
Abstract
Ischemic stroke is a major cause of morbidity and mortality, yet lacks effective neuroprotective treatments. The aim of this work was to investigate whether treatment with isorhamnetin protected the brain against ischemic injury in mice. Experimental stroke mice underwent the filament model of middle cerebral artery occlusion with reperfusion. Treatment with isorhamnetin or vehicle was initiated immediately at the onset of reperfusion. It was found that treatment of experimental stroke mice with isorhamnetin reduced infarct volume and caspase-3 activity (a biomarker of apoptosis), and improved neurological function recovery. Treatment of experimental stroke mice with isorhamnetin attenuated cerebral edema, improved blood-brain barrier function, and upregulated gene expression of tight junction proteins including occludin, ZO-1, and claudin-5. Treatment of experimental stroke mice with isorhamnetin activated Nrf2/HO-1, suppressed iNOS/NO, and led to reduced formation of MDA and 3-NT in ipsilateral cortex. In addition, treatment of experimental stroke mice with isorhamnetin suppressed activity of MPO (a biomarker of neutrophil infiltration) and reduced protein levels of IL-1β, IL-6, and TNF-α in ipsilateral cortex. Furthermore, it was found that treatment of experimental stroke mice with isorhamnetin reduced mRNA and protein expression of NMDA receptor subunit NR1 in ipsilateral cortex. In conclusion, treatment with isorhamnetin protected the brain against ischemic injury in mice. Isorhamnetin could thus be envisaged as a countermeasure for ischemic stroke but remains to be tested in humans.
Collapse
Affiliation(s)
- Jin-Jing Zhao
- Department of Neurology, 305th Hospital of the People's Liberation Army, Jia13 Wenjin Road, Beijing, 100017, China
| | - Jin-Qing Song
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Shu-Yi Pan
- Department of Hyperbaric Oxygen, Navy General Hospital of People's Liberation Army, Beijing, 100048, China
| | - Kai Wang
- Department of Neurology, 305th Hospital of the People's Liberation Army, Jia13 Wenjin Road, Beijing, 100017, China.
| |
Collapse
|
35
|
Zeynalov E, Jones SM. Recent advances and future directions in preclinical research of arginine-vasopressin (AVP) receptor blocker conivaptan in the context of stroke. Neural Regen Res 2016; 11:392-3. [PMID: 27127464 PMCID: PMC4828990 DOI: 10.4103/1673-5374.179038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Emil Zeynalov
- Swedish Medical Center, Neurotrauma Research Department Englewood, Englewood, CO, USA
| | - Susan M Jones
- Swedish Medical Center, Neurotrauma Research Department Englewood, Englewood, CO, USA
| |
Collapse
|
36
|
Jia SW, Liu XY, Wang SC, Wang YF. Vasopressin Hypersecretion-Associated Brain Edema Formation in Ischemic Stroke: Underlying Mechanisms. J Stroke Cerebrovasc Dis 2016; 25:1289-300. [PMID: 27068863 DOI: 10.1016/j.jstrokecerebrovasdis.2016.02.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/21/2016] [Accepted: 02/01/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Brain edema formation is a major cause of brain damages and the high mortality of ischemic stroke. The aim of this review is to explore the relationship between ischemic brain edema formation and vasopressin (VP) hypersecretion in addition to the oxygen and glucose deprivation and the ensuing reperfusion injury. METHODS Pertinent studies involving ischemic stroke, brain edema formation, astrocytes, and VP were identified by a search of the PubMed and the Web of Science databases in January 2016. Based on clinical findings and reports of animal experiments using ischemic stroke models, this systematic review reanalyzes the implication of individual reports in the edema formation and then establishes the inherent links among them. RESULTS This systematic review reveals that cytotoxic edema and vasogenic brain edema in classical view are mainly under the influence of a continuous malfunction of astrocytic plasticity. Adaptive VP secretion can modulate membrane ion transport, water permeability, and blood-brain barrier integrity, which are largely via changing astrocytic plasticity. Maladaptive VP hypersecretion leads to disruptions of ion and water balance across cell membranes as well as the integrity of the blood-brain barrier. This review highlights our current understandings of the cellular mechanisms underlying ischemic brain edema formation and its association with VP hypersecretion. CONCLUSIONS VP hypersecretion promotes brain edema formation in ischemic stroke by disrupting hydromineral balance in the neurovascular unit; suppressing VP hypersecretion has the potential to alleviate ischemic brain edema.
Collapse
Affiliation(s)
- Shu-Wei Jia
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Xiao-Yu Liu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Stephani C Wang
- Department of Surgery, Albany Medical Center, Albany, New York
| | - Yu-Feng Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China.
| |
Collapse
|
37
|
Abstract
Arginine vasopressin (AVP) plays a major role in the homeostasis of fluid balance, vascular tonus, and the regulation of the endocrine stress response. The measurement of AVP levels is difficult due to its short half-life and laborious method of detection. Copeptin is a more stable peptide derived from the same precursor molecule, is released in an equimolar ratio to AVP, and has a very similar response to osmotic, hemodynamic, and stress-related stimuli. In fact, copeptin has been propagated as surrogate marker to indirectly determine circulating AVP concentrations in various conditions. Here, we present an overview of the current knowledge on AVP and copeptin in perinatology with a particular focus on the baby's transition from placenta to lung breathing. We performed a systematic review of the literature on fetal stress hormone levels, including norepinephrine, cortisol, AVP, and copeptin, in regard to birth stress. Finally, diagnostic and therapeutic options for copeptin measurement and AVP functions are discussed.
Collapse
Affiliation(s)
- Katrina Suzanne Evers
- Division of Neonatology, University of Basel Children's Hospital (UKBB) , Basel , Switzerland
| | - Sven Wellmann
- Division of Neonatology, University of Basel Children's Hospital (UKBB) , Basel , Switzerland
| |
Collapse
|