1
|
Peng B, Mohammed FS, Tang X, Liu J, Sheth KN, Zhou J. Nanotechnology approaches to drug delivery for the treatment of ischemic stroke. Bioact Mater 2025; 43:145-161. [PMID: 39386225 PMCID: PMC11462157 DOI: 10.1016/j.bioactmat.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024] Open
Abstract
Ischemic stroke is a major global public health concern that lacks effective treatment options. A significant challenge lies in delivering therapeutic agents to the brain due to the restrictive nature of the blood-brain barrier (BBB). The BBB's selectivity hampers the delivery of therapeutically relevant quantities of agents to the brain, resulting in a lack of FDA-approved pharmacotherapies for stroke. In this article, we review therapeutic agents that have been evaluated in clinical trials or are currently undergoing clinical trials. Subsequently, we survey strategies for synthesizing and engineering nanoparticles (NPs) for drug delivery to the ischemic brain. We then provide insights into the potential clinical translation of nanomedicine, offering a perspective on its transformative role in advancing stroke treatment strategies. In summary, existing literature suggests that drug delivery represents a major barrier for clinical translation of stroke pharmacotherapies. While nanotechnology has shown significant promise in addressing this challenge, further advancements aimed at improving delivery efficiency and simplifying formulations are necessary for successful clinical translation.
Collapse
Affiliation(s)
- Bin Peng
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Farrah S. Mohammed
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Biomedical Engineering, New Haven, CT, 06510, USA
| | - Xiangjun Tang
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Neurosurgery, Taihe Hospital, Hubei, 442000, PR China
| | - Jia Liu
- Department of Neurosurgery, New Haven, CT, 06510, USA
| | - Kevin N. Sheth
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Neurology, Yale University, New Haven, CT, 06510, USA
| | - Jiangbing Zhou
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Biomedical Engineering, New Haven, CT, 06510, USA
| |
Collapse
|
2
|
Díaz-Pérez A, Pérez B, Manich G, García-Aranda J, Navarro X, Penas C, Jiménez-Altayó F. Histone deacetylase inhibition by suberoylanilide hydroxamic acid during reperfusion promotes multifaceted brain and vascular protection in spontaneously hypertensive rats with transient ischaemic stroke. Biomed Pharmacother 2024; 172:116287. [PMID: 38382328 DOI: 10.1016/j.biopha.2024.116287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/07/2024] [Accepted: 02/17/2024] [Indexed: 02/23/2024] Open
Abstract
Hypertension is the most prevalent modifiable risk factor for stroke and is associated with worse functional outcomes. Pharmacological inhibition of histone deacetylases by suberoylanilide hydroxamic acid (SAHA) modulates gene expression and has emerged as a promising therapeutic approach to reduce ischaemic brain injury. Here, we have tested the therapeutic potential of SAHA administered during reperfusion in adult male spontaneously hypertensive (SHR) rats subjected to transient middle cerebral artery occlusion (tMCAO; 90 min occlusion/24 h reperfusion). Animals received a single dose of SAHA (50 mg/kg) or vehicle i.p. at 1, 4, or 6 h after reperfusion onset. The time-course of brain histone H3 acetylation was studied. After tMCAO, drug brain penetrance and beneficial effects on behavioural outcomes, infarct volume, oedema, angiogenesis, blood-brain barrier integrity, cerebral artery oxidative stress and remodelling, and brain and vascular inflammation were evaluated. SAHA increased brain histone H3 acetylation from 1 to 6 h after injection, reaching the ischaemic brain administered during reperfusion. Treatment given at 4 h after reperfusion onset improved neurological score, reduced infarct volume and oedema, attenuated microglial activation, prevented exacerbated MCA angiogenic sprouting and blood-brain barrier breakdown, normalised MCA oxidative stress and remodelling, and modulated brain and cerebrovascular cytokine expression. Overall, we demonstrate that SAHA administered during early reperfusion exerts robust brain and vascular protection after tMCAO in hypertensive rats. These findings are aligned with previous research in ischaemic normotensive mice and help pave the way to optimise the design of clinical trials assessing the effectiveness and safety of SAHA in ischaemic stroke.
Collapse
Affiliation(s)
- Andrea Díaz-Pérez
- Department of Pharmacology, Therapeutic and Toxicology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Belén Pérez
- Department of Pharmacology, Therapeutic and Toxicology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Gemma Manich
- Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Human Anatomy and Embriology Unit, Department of Morphological Sciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Julián García-Aranda
- Department of Pharmacology, Therapeutic and Toxicology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Xavier Navarro
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Red Española de Terapias Avanzadas (RED-TERAV), Instituto de Salud Carlos III, Madrid, Spain
| | - Clara Penas
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Red Española de Terapias Avanzadas (RED-TERAV), Instituto de Salud Carlos III, Madrid, Spain.
| | - Francesc Jiménez-Altayó
- Department of Pharmacology, Therapeutic and Toxicology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
3
|
Leira EC, Planas AM, Chauhan AK, Chamorro A. Uric Acid: A Translational Journey in Cerebroprotection That Spanned Preclinical and Human Data. Neurology 2023; 101:1068-1074. [PMID: 37848338 PMCID: PMC10752646 DOI: 10.1212/wnl.0000000000207825] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/12/2023] [Indexed: 10/19/2023] Open
Abstract
Uric acid (UA) is a strong endogenous antioxidant that neutralizes the toxicity of peroxynitrite and other reactive species on the neurovascular unit generated during and after acute brain ischemia. The realization that a rapid reduction of UA levels during an acute ischemic stroke was associated with a worse stroke outcome paved the way to investigate the value of exogenous UA supplementation to counteract the progression of redox-mediated ischemic brain damage. The long translational journey for UA supplementation recently reached a critical milestone when the results of the multicenter NIH stroke preclinical assessment network (SPAN) were reported. In a novel preclinical paradigm, 6 treatment candidates including UA supplementation were selected and tested in 6 independent laboratories following predefined criteria and strict methodological rigor. UA supplementation was the only intervention in SPAN that exceeded the prespecified efficacy boundary with male and female animals, young mice, young rats, aging mice, obese mice, and spontaneously hypertensive rats. This unprecedented achievement will allow UA to undergo clinical testing in a pivotal clinical trial through a NIH StrokeNet thrombectomy endovascular platform created to assess new treatment strategies in patients treated with mechanical thrombectomy. UA is a particularly appealing adjuvant intervention for mechanical thrombectomy because it targets the microcirculatory hypoperfusion and oxidative stress that limits the efficacy of this therapy. This descriptive review aims to summarize the translational development of UA supplementation, highlighting those aspects that likely contributed to its success. It includes having a well-defined target and mechanism of action, and an approach that simultaneously integrated rigorous preclinical assessment, with epidemiologic and preliminary human intervention studies. Validation of the clinical value of UA supplementation in a pivotal trial would confirm the translational value of the SPAN paradigm in preclinical research.
Collapse
Affiliation(s)
- Enrique C Leira
- From the Department of Neurology (E.L., A.C.), and Departments of Neurosurgery & Epidemiology (E.L.), University of Iowa, Iowa City; Institute of Biomedical Research of Barcelona (IIBB) (A.M.P.), Spanish National Research Council (CSIC); August Pi i Sunyer Biomedical Research Institute (IDIBAPS) (A.M.P., A.C.), Barcelona, Spain; Department of Internal Medicine (A.K.C.), University of Iowa, Iowa City; and Hospital Clinic (A.C.), University of Barcelona, Spain
| | - Anna M Planas
- From the Department of Neurology (E.L., A.C.), and Departments of Neurosurgery & Epidemiology (E.L.), University of Iowa, Iowa City; Institute of Biomedical Research of Barcelona (IIBB) (A.M.P.), Spanish National Research Council (CSIC); August Pi i Sunyer Biomedical Research Institute (IDIBAPS) (A.M.P., A.C.), Barcelona, Spain; Department of Internal Medicine (A.K.C.), University of Iowa, Iowa City; and Hospital Clinic (A.C.), University of Barcelona, Spain
| | - Anil K Chauhan
- From the Department of Neurology (E.L., A.C.), and Departments of Neurosurgery & Epidemiology (E.L.), University of Iowa, Iowa City; Institute of Biomedical Research of Barcelona (IIBB) (A.M.P.), Spanish National Research Council (CSIC); August Pi i Sunyer Biomedical Research Institute (IDIBAPS) (A.M.P., A.C.), Barcelona, Spain; Department of Internal Medicine (A.K.C.), University of Iowa, Iowa City; and Hospital Clinic (A.C.), University of Barcelona, Spain
| | - Angel Chamorro
- From the Department of Neurology (E.L., A.C.), and Departments of Neurosurgery & Epidemiology (E.L.), University of Iowa, Iowa City; Institute of Biomedical Research of Barcelona (IIBB) (A.M.P.), Spanish National Research Council (CSIC); August Pi i Sunyer Biomedical Research Institute (IDIBAPS) (A.M.P., A.C.), Barcelona, Spain; Department of Internal Medicine (A.K.C.), University of Iowa, Iowa City; and Hospital Clinic (A.C.), University of Barcelona, Spain.
| |
Collapse
|
4
|
Feng J, Zheng Y, Guo M, Ares I, Martínez M, Lopez-Torres B, Martínez-Larrañaga MR, Wang X, Anadón A, Martínez MA. Oxidative stress, the blood-brain barrier and neurodegenerative diseases: The critical beneficial role of dietary antioxidants. Acta Pharm Sin B 2023; 13:3988-4024. [PMID: 37799389 PMCID: PMC10547923 DOI: 10.1016/j.apsb.2023.07.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/16/2023] [Accepted: 06/13/2023] [Indexed: 10/07/2023] Open
Abstract
In recent years, growing awareness of the role of oxidative stress in brain health has prompted antioxidants, especially dietary antioxidants, to receive growing attention as possible treatments strategies for patients with neurodegenerative diseases (NDs). The most widely studied dietary antioxidants include active substances such as vitamins, carotenoids, flavonoids and polyphenols. Dietary antioxidants are found in usually consumed foods such as fresh fruits, vegetables, nuts and oils and are gaining popularity due to recently growing awareness of their potential for preventive and protective agents against NDs, as well as their abundant natural sources, generally non-toxic nature, and ease of long-term consumption. This review article examines the role of oxidative stress in the development of NDs, explores the 'two-sidedness' of the blood-brain barrier (BBB) as a protective barrier to the nervous system and an impeding barrier to the use of antioxidants as drug medicinal products and/or dietary antioxidants supplements for prevention and therapy and reviews the BBB permeability of common dietary antioxidant suplements and their potential efficacy in the prevention and treatment of NDs. Finally, current challenges and future directions for the prevention and treatment of NDs using dietary antioxidants are discussed, and useful information on the prevention and treatment of NDs is provided.
Collapse
Affiliation(s)
- Jin Feng
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
| | - Youle Zheng
- MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan 430070, China
| | - Mingyue Guo
- MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan 430070, China
| | - Irma Ares
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - Marta Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - Bernardo Lopez-Torres
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - María-Rosa Martínez-Larrañaga
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - Arturo Anadón
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| | - María-Aránzazu Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), And Research Institute Hospital 12 de Octubre (i+12), Madrid 28040, Spain
| |
Collapse
|
5
|
Deep Learning-Based Computed Tomography Perfusion Imaging to Evaluate the Effectiveness and Safety of Thrombolytic Therapy for Cerebral Infarct with Unknown Time of Onset. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:9684584. [PMID: 35615733 PMCID: PMC9110226 DOI: 10.1155/2022/9684584] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/03/2022] [Accepted: 04/08/2022] [Indexed: 12/30/2022]
Abstract
This study was aimed to discuss the effectiveness and safety of deep learning-based computed tomography perfusion (CTP) imaging in the thrombolytic therapy for acute cerebral infarct with unknown time of onset. A total of 100 patients with acute cerebral infarct with unknown time of onset were selected as the research objects. All patients received thrombolytic therapy. According to different image processing methods, they were divided into the algorithm group (artificial intelligence algorithm-based image processing group) and the control group (conventional method-based image processing group). After that, the evaluations of effectiveness and safety of thrombolytic therapy for the patients with acute cerebral infarct in the two groups were compared. The research results demonstrated that artificial intelligence algorithm-based CTP imaging showed significant diagnostic effects and the image quality in the algorithm group was remarkably higher than that in the control group (P < 0.05). Besides, the overall image quality of algorithm group was relatively higher. The differences in the National Institute of Health stroke scale (NIHSS) scores for the two groups indicated that the thrombolytic effect on the algorithm group was superior to that on the control group. Thrombolytic therapy for the algorithm group showed therapeutic effects on neurologic impairment. The symptomatic intracranial hemorrhage rate of the algorithm group within 24 hours was lower than the hemorrhage conversion rate of the control group, and the difference between the two groups was 14%. The data differences between the two groups showed statistical significance (P < 0.05). The results demonstrated that the safety of guided thrombolytic therapy for the algorithm group was higher than that in the control group. To sum up, deep learning-based CTP images showed the clinical application values in the diagnosis of cerebral infarct.
Collapse
|
6
|
Liu Y, Liu X, Jia J, Guo J, Li G, Zhao X. Uric Acid and Clinical Outcomes in Young Patients with Ischemic Stroke. Neuropsychiatr Dis Treat 2022; 18:2219-2228. [PMID: 36199274 PMCID: PMC9529006 DOI: 10.2147/ndt.s373493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/14/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND AND PURPOSE There is limited available evidence for the relationship between uric acid (UA) levels and ischemic stroke in young adults. We aimed to explore the association between UA levels and acute ischemic stroke (AIS) in young patients. MATERIALS AND METHODS This was a prospective and observational study. We recruited young patients aged 18-45 years with AIS at our tertiary hospital. Patients were categorized into four groups according to quartiles of UA levels. The primary outcome was functional outcome at 3 months. The secondary outcomes included stroke severity, in-hospital complications, and functional outcome at discharge. Modified Rankin Scale (mRS) scores were used to assess functional outcome as poor (mRS=2-6) or favorable(mRS=0-1). RESULTS A total of 636 patients were enrolled in the current analysis. The four groups were defined as follows: Q1≤289.8 µmol/L, 289.8 µmol/ L<Q2≤349.0 µmol/L, 349.0 µmol/L<Q3≤421 µmol/L, and Q4>421 µmol/L. Multiple logistic regression analysis showed that UA levels were not significantly predictive of functional outcome either at discharge or at 3 months after AIS. However, compared to Q1, higher UA levels were significantly negatively associated with the rate of moderate-severe stroke (NIHSS≥5) at admission (p for trend =0.016). Furthermore, a reduction in the risk for in-hospital pneumonia was significantly associated with higher UA levels compared to Q1 (P for trend < 0.0001). CONCLUSION Serum UA was a protective factor for stroke severity and in-hospital pneumonia after AIS in young patients. However, we were unable to identify the predictive significance of UA for functional outcome either at discharge or at 3 months after AIS.
Collapse
Affiliation(s)
- Yanfang Liu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.,China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China.,Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Xinmin Liu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.,China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China
| | - Jiaokun Jia
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.,China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China.,Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Jiahuan Guo
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.,China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China
| | - Guangshuo Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.,China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China
| | - Xingquan Zhao
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.,China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China.,Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, People's Republic of China.,Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
7
|
Aliena-Valero A, Baixauli-Martín J, Castelló-Ruiz M, Torregrosa G, Hervás D, Salom JB. Effect of uric acid in animal models of ischemic stroke: A systematic review and meta-analysis. J Cereb Blood Flow Metab 2021; 41:707-722. [PMID: 33210575 PMCID: PMC7983496 DOI: 10.1177/0271678x20967459] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Addition of uric acid (UA) to thrombolytic therapy, although safe, showed limited efficacy in improving patients' stroke outcome, despite alleged neuroprotective effects of UA in preclinical research. This systematic review assessed the effects of UA on brain structural and functional outcomes in animal models of ischemic stroke. We searched Medline, Embase and Web of Science to identify 16 and 14 eligible rodent studies for qualitative and quantitative synthesis, respectively. Range of evidence met 10 of a possible 13 STAIR criteria. Median (Q1, Q3) quality score was 7.5 (6, 10) on the CAMARADES 15-item checklist. For each outcome, we used standardised mean difference (SMD) as effect size and random-effects modelling. Meta-analysis showed that UA significantly reduced infarct size (SMD: -1.18; 95% CI [-1.47, -0.88]; p < 0.001), blood-brain barrier (BBB) impairment/oedema (SMD: -0.72; 95% CI [-0.97, -0.48]; p < 0.001) and neurofunctional deficit (SMD: -0.98; 95% CI [-1.32, -0.63]; p < 0.001). Overall, there was low to moderate between-study heterogeneity and sizeable publication bias. In conclusion, published rodent data suggest that UA improves outcome following ischemic stroke by reducing infarct size, improving BBB integrity and ameliorating neurofunctional condition. Specific recommendations are given for further high-quality preclinical research required to better inform clinical research.
Collapse
Affiliation(s)
- Alicia Aliena-Valero
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universidad de Valencia, Valencia, Spain
| | | | - María Castelló-Ruiz
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universidad de Valencia, Valencia, Spain.,Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, Valencia, Spain
| | - Germán Torregrosa
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universidad de Valencia, Valencia, Spain
| | - David Hervás
- Unidad de Bioestadística, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Juan B Salom
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universidad de Valencia, Valencia, Spain.,Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| |
Collapse
|
8
|
Aliena-Valero A, Rius-Pérez S, Baixauli-Martín J, Torregrosa G, Chamorro Á, Pérez S, Salom JB. Uric Acid Neuroprotection Associated to IL-6/STAT3 Signaling Pathway Activation in Rat Ischemic Stroke. Mol Neurobiol 2021; 58:408-423. [PMID: 32959172 DOI: 10.1007/s12035-020-02115-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 09/02/2020] [Indexed: 12/21/2022]
Abstract
Despite the promising neuroprotective effects of uric acid (UA) in acute ischemic stroke, the seemingly pleiotropic underlying mechanisms are not completely understood. Recent evidence points to transcription factors as UA targets. To gain insight into the UA mechanism of action, we investigated its effects on pertinent biomarkers for the most relevant features of ischemic stroke pathophysiology: (1) oxidative stress (antioxidant enzyme mRNAs and MDA), (2) neuroinflammation (cytokine and Socs3 mRNAs, STAT3, NF-κB p65, and reactive microglia), (3) brain swelling (Vegfa, Mmp9, and Timp1 mRNAs), and (4) apoptotic cell death (Bcl-2, Bax, caspase-3, and TUNEL-positive cells). Adult male Wistar rats underwent intraluminal filament transient middle cerebral artery occlusion (tMCAO) and received UA (16 mg/kg) or vehicle (Locke's buffer) i.v. at 20 min reperfusion. The outcome measures were neurofunctional deficit, infarct, and edema. UA treatment reduced cortical infarct and brain edema, as well as neurofunctional impairment. In brain cortex, increased UA: (1) reduced tMCAO-induced increases in Vegfa and Mmp9/Timp1 ratio expressions; (2) induced Sod2 and Cat expressions and reduced MDA levels; (3) induced Il6 expression, upregulated STAT3 and NF-κB p65 phosphorylation, induced Socs3 expression, and inhibited microglia activation; and (4) ameliorated the Bax/Bcl-2 ratio and induced a reduction in caspase-3 cleavage as well as in TUNEL-positive cell counts. In conclusion, the mechanism for morphological and functional neuroprotection by UA in ischemic stroke is multifaceted, since it is associated to activation of the IL-6/STAT3 pathway, attenuation of edematogenic VEGF-A/MMP-9 signaling, and modulation of relevant mediators of oxidative stress, neuroinflammation, and apoptotic cell death.
Collapse
Affiliation(s)
- Alicia Aliena-Valero
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universitat de València, Torre A, Lab 5.05, Ave Fernando Abril Martorell 106, 46026, Valencia, Spain
- Departamento de Fisiología, Facultad de Farmacia, Universitat de València, Ave Vicent Andrés Estellés s/n, Burjassot, 46100, Valencia, Spain
| | - Sergio Rius-Pérez
- Departamento de Fisiología, Facultad de Farmacia, Universitat de València, Ave Vicent Andrés Estellés s/n, Burjassot, 46100, Valencia, Spain
| | - Júlia Baixauli-Martín
- Departamento de Fisiología, Facultad de Farmacia, Universitat de València, Ave Vicent Andrés Estellés s/n, Burjassot, 46100, Valencia, Spain
| | - Germán Torregrosa
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universitat de València, Torre A, Lab 5.05, Ave Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Ángel Chamorro
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Comprehensive Stroke Center, Department of Neuroscience, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Departamento de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Salvador Pérez
- Departamento de Fisiología, Facultad de Farmacia, Universitat de València, Ave Vicent Andrés Estellés s/n, Burjassot, 46100, Valencia, Spain.
| | - Juan B Salom
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universitat de València, Torre A, Lab 5.05, Ave Fernando Abril Martorell 106, 46026, Valencia, Spain.
- Departamento de Fisiología, Facultad de Farmacia, Universitat de València, Ave Vicent Andrés Estellés s/n, Burjassot, 46100, Valencia, Spain.
| |
Collapse
|
9
|
Stenosis coexists with compromised α1-adrenergic contractions in the ascending aorta of a mouse model of Williams-Beuren syndrome. Sci Rep 2020; 10:889. [PMID: 31965005 PMCID: PMC6972706 DOI: 10.1038/s41598-020-57803-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 01/07/2020] [Indexed: 11/08/2022] Open
Abstract
Williams-Beuren syndrome (WBS) is a rare disorder caused by a heterozygous deletion of 26–28 contiguous genes that affects the brain and cardiovascular system. Here, we investigated whether WBS affects aortic structure and function in the complete deletion (CD) mouse model harbouring the most common deletion found in WBS patients. Thoracic aortas from 3–4 months-old male CD mice and wild-type littermates were mounted in wire myographs or were processed for histomorphometrical analysis. Nitric oxide synthase (NOS) isoforms and oxidative stress levels were assessed. Ascending aortas from young adult CD mice showed moderate (50%) luminal stenosis, whereas endothelial function and oxidative stress were comparable to wild-type. CD mice showed greater contractions to KCl. However, α1-adrenergic contractions to phenylephrine, but not with a thromboxane analogue, were compromised. Decreased phenylephrine responses were not affected by selective inducible NOS blockade with 1400 W, but were prevented by the non-selective NOS inhibitor L-NAME and the selective neuronal NOS inhibitor SMTC. Consistently, CD mice showed increased neuronal NOS expression in aortas. Overall, aortic stenosis in CD mice coexists with excessive nNOS-derived NO signaling that compromises ascending aorta α1-adrenergic contractions. We suggest that increased neuronal NOS signaling may act as a physiological ‘brake’ against the detrimental effects of stenosis.
Collapse
|
10
|
Zuo G, Zhang T, Huang L, Araujo C, Peng J, Travis Z, Okada T, Ocak U, Zhang G, Tang J, Lu X, Zhang JH. Activation of TGR5 with INT-777 attenuates oxidative stress and neuronal apoptosis via cAMP/PKCε/ALDH2 pathway after subarachnoid hemorrhage in rats. Free Radic Biol Med 2019; 143:441-453. [PMID: 31493504 PMCID: PMC6848789 DOI: 10.1016/j.freeradbiomed.2019.09.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/27/2019] [Accepted: 09/03/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Oxidative stress and neuronal apoptosis play important roles in the pathogenesis of early brain injury (EBI) after subarachnoid hemorrhage (SAH). The activation of TGR5, a novel membrane-bound bile acid receptor, possesses anti-oxidative stress and anti-apoptotic effects in hepatobiliary disease and kidney disease. The present study aimed to explore the neuroprotective effect of TGR5 activation against EBI after SAH and the potential underlying mechanisms. METHODS The endovascular perforation model of SAH was performed on 199 Sprague Dawley rats to investigate the beneficial effects of TGR5 activation after SAH. INT-777, a specific synthetic TGR5 agonist, was administered intranasally at 1 h after SAH induction. TGR5 CRISPR and ALDH2 CRISPR were administered intracerebroventricularly at 48 h before SAH to illuminate potential mechanisms. The SAH grade, short-term and long-term neurobehavioral tests, TUNEL staining, Fluoro-Jade C staining, Nissl staining, immunofluorescence staining, and western blots were performed at 24 h after SAH. RESULTS The expressions of endogenous TGR5 and ALDH2 gradually increased and peaked at 24 h after SAH. TGR5 was expressed primarily in neurons, as well as in astrocytes and microglia. The activation of TGR5 with INT-777 significantly improved the short-term and long-term neurological deficits, accompanied by reduced the oxidative stress and neuronal apoptosis at 24 h after SAH. Moreover, INT-777 treatment significantly increased the expressions of TGR5, cAMP, phosphorylated PKCε, ALDH2, HO-1, and Bcl-2, while downregulated the expressions of 4-HNE, Bax, and Cleaved Caspase-3. TGR5 CRISPR and ALDH2 CRISPR abolished the neuroprotective effects of TGR5 activation after SAH. CONCLUSIONS In summary, the activation of TGR5 with INT-777 attenuated oxidative stress and neuronal apoptosis via the cAMP/PKCε/ALDH2 signaling pathway after SAH in rats. Furthermore, TGR5 may serve as a novel therapeutic target to ameliorate EBI after SAH.
Collapse
Affiliation(s)
- Gang Zuo
- Department of Neurosurgery, The Affiliated Taicang Hospital, Soochow University, Taicang, Suzhou, Jiangsu, 215400, China; Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Tongyu Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA; Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, China
| | - Lei Huang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA; Department of Neurosurgery, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Camila Araujo
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Jun Peng
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Zachary Travis
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Takeshi Okada
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Umut Ocak
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Guangyu Zhang
- Mass Spectrometry Core Facility, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Xiaojun Lu
- Department of Neurosurgery, The Affiliated Taicang Hospital, Soochow University, Taicang, Suzhou, Jiangsu, 215400, China.
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA; Department of Neurosurgery, Loma Linda University, Loma Linda, CA, 92350, USA; Department of Anesthesiology, Loma Linda University, Loma Linda, CA, 92350, USA.
| |
Collapse
|
11
|
Amaro S, Jiménez-Altayó F, Chamorro Á. Uric acid therapy for vasculoprotection in acute ischemic stroke. Brain Circ 2019; 5:55-61. [PMID: 31334357 PMCID: PMC6611195 DOI: 10.4103/bc.bc_1_19] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/18/2019] [Accepted: 05/02/2019] [Indexed: 02/07/2023] Open
Abstract
Uric acid (UA) is a product of the catabolism of purine nucleotides, the principal constituents of DNA, RNA, and cellular energy stores, such as adenosine triphosphate. The main properties of UA include scavenging of hydroxyl radicals, superoxide anion, hydrogen peroxide, and peroxynitrite that make this compound to be the most potent antioxidant in the human plasma. As the result of two silencing mutations in the gene of the hepatic enzyme uricase which degrades UA to allantoin, humans have higher levels of UA than most mammals. However, these levels rapidly decrease following an acute ischemic stroke (AIS), and this decrement has been associated to worse stroke outcomes. This review highlights the safety and potential clinical value of UA therapy in AIS, particularly in patients more exposed to redox-mediated mechanism following the onset of ischemia, such as women, hyperglycemic patients, or patients treated with mechanical thrombectomy. The clinical findings are supported by preclinical data gathered in different laboratories, and in assorted animal species which include male and female individuals or animals harboring comorbidities frequently encountered in patients with AIS, such as hyperglycemia or hypertension. A remarkable finding in these studies is that UA targets its main effects in the brain vasculature since available evidence suggests that does not seem to cross the blood–brain barrier. Altogether, the available data with UA therapy extend the importance of vasculoprotection for effective neuroprotection at the bedside and reinforce the role of endothelial cells after brain ischemia for an increased survival of the whole neurovascular unit.
Collapse
Affiliation(s)
- Sergi Amaro
- Comprehensive Stroke Center, Hospital Clínic, University of Barcelona, Barcelona, Spain.,Department of Neuroscience, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francesc Jiménez-Altayó
- Department de Farmacologia, de Terapèutica i de Toxicologia, Facultat de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ángel Chamorro
- Comprehensive Stroke Center, Hospital Clínic, University of Barcelona, Barcelona, Spain.,Department of Neuroscience, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
12
|
Uric acid treatment after stroke modulates the Krüppel-like factor 2-VEGF-A axis to protect brain endothelial cell functions: Impact of hypertension. Biochem Pharmacol 2019; 164:115-128. [DOI: 10.1016/j.bcp.2019.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/03/2019] [Indexed: 12/29/2022]
|
13
|
Gandhi R, Tsoumpas C. Preclinical Imaging Biomarkers for Postischaemic Neurovascular Remodelling. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:3128529. [PMID: 30863220 PMCID: PMC6378027 DOI: 10.1155/2019/3128529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/22/2018] [Accepted: 12/04/2018] [Indexed: 11/30/2022]
Abstract
In the pursuit of understanding the pathological alterations that underlie ischaemic injuries, such as vascular remodelling and reorganisation, there is a need for recognising the capabilities and limitations of in vivo imaging techniques. Thus, this review presents contemporary published research of imaging modalities that have been implemented to study postischaemic neurovascular changes in small animals. A comparison of the technical aspects of the various imaging tools is included to set the framework for identifying the most appropriate methods to observe postischaemic neurovascular remodelling. A systematic search of the PubMed® and Elsevier's Scopus databases identified studies that were conducted between 2008 and 2018 to explore postischaemic neurovascular remodelling in small animal models. Thirty-five relevant in vivo imaging studies are included, of which most made use of magnetic resonance imaging or positron emission tomography, whilst various optical modalities were also utilised. Notably, there is an increasing trend of using multimodal imaging to exploit the most beneficial properties of each imaging technique to elucidate different aspects of neurovascular remodelling. Nevertheless, there is still scope for further utilising noninvasive imaging tools such as contrast agents or radiotracers, which will have the ability to monitor neurovascular changes particularly during restorative therapy. This will facilitate more successful utility of the clinical imaging techniques in the interpretation of neurovascular reorganisation over time.
Collapse
Affiliation(s)
- Richa Gandhi
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9NL, West Yorkshire, UK
| | - Charalampos Tsoumpas
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9NL, West Yorkshire, UK
| |
Collapse
|