1
|
YAMANE YUI, LI XIAOJIA, HANAFUSA KEI, NAKAYAMA HITOSHI, WATANABE KOJI, IWABUCHI KAZUHISA, HAYASHIDA MASAKAZU. Dexmedetomidine Pretreatment of Neuronal Cells Has Protective Effect Against Cell Death During Oxygen-glucose Deprivation/Reoxygenation, Based on IGF-1 Production. JUNTENDO IJI ZASSHI = JUNTENDO MEDICAL JOURNAL 2024; 70:360-367. [PMID: 39545230 PMCID: PMC11560337 DOI: 10.14789/jmj.jmj23-0037-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 06/10/2024] [Indexed: 11/17/2024]
Abstract
Objective Insulin-like growth factor 1 (IGF-1) protects neuronal-cell damage by ischemia. Although neuronal cells have been reported to produce IGF-1, the molecular mechanisms remains obscure. Dexmedetomidine (DEX) protects neuronal cells from ischemic damage. We investigated the involvement of IGF-1 in the effect of DEX pretreatment on neuronal ischemic damage using an in vitro mouse hippocampal neuron model. Materials We used Dexmedetomidine and cryopreserved passaged mouse hippocampal neuronal HT22. Other reagents in this study were analytical grade. Methods Ischemia-reperfusion was modeled using the in vitro oxygen-glucose deprivation/reoxygenation (OGD/R). The effect of DEX was examined by incubating cells in DEX-containing medium for 1 hour prior to OGD/R. The cell damages were evaluated by lactate dehydrogenase (LDH) release. The amount of released IGF-1 were evaluated quantitatively by ELISA. The degree of Akt phosphorylation was evaluated by western blotting. Results OGD/R loading promoted LDH release from neuronal cells, while DEX pretreatment suppressed the LDH release. IGF-1 release from them was primed by DEX pretreatment under OGD/R condition, but not under normal conditions. Akt was activated in DEX-pretreated cells following OGD/R loading. IGF-1 neutralizing antibody (αIGF-1) eliminated the above effects of DEX pretreatment. However, IGF-1 receptor expression in neuronal cells was not affected by DEX pretreatment prior to OGD/R loading. Conclusions Our results demonstrate that neuronal cells primed with DEX under OGD/R conditions could release IGF-1 and potentially protect themselves via the IGF-1/Akt pathway. Consequently, it appears that neuronal cells activated by DEX have the capacity to self-protect from ischemic damage.
Collapse
Affiliation(s)
| | | | | | | | | | - KAZUHISA IWABUCHI
- Corresponding author: Kazuhisa Iwabuchi, Infection Control Nursing, Juntendo University Graduate School of Health Care and Nursing, 2-5-1 Takasu, Urayasu-shi, Chiba 279-0023, Japan, TEL: +81-47-353-3171 E-mail:
| | | |
Collapse
|
2
|
Chen R, Wang X, Li N, Golubnitschaja O, Zhan X. Body fluid multiomics in 3PM-guided ischemic stroke management: health risk assessment, targeted protection against health-to-disease transition, and cost-effective personalized approach are envisaged. EPMA J 2024; 15:415-452. [PMID: 39239108 PMCID: PMC11371995 DOI: 10.1007/s13167-024-00376-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024]
Abstract
Because of its rapid progression and frequently poor prognosis, stroke is the third major cause of death in Europe and the first one in China. Many independent studies demonstrated sufficient space for prevention interventions in the primary care of ischemic stroke defined as the most cost-effective protection of vulnerable subpopulations against health-to-disease transition. Although several studies identified molecular patterns specific for IS in body fluids, none of these approaches has yet been incorporated into IS treatment guidelines. The advantages and disadvantages of individual body fluids are thoroughly analyzed throughout the paper. For example, multiomics based on a minimally invasive approach utilizing blood and its components is recommended for real-time monitoring, due to the particularly high level of dynamics of the blood as a body system. On the other hand, tear fluid as a more stable system is recommended for a non-invasive and patient-friendly holistic approach appropriate for health risk assessment and innovative screening programs in cost-effective IS management. This article details aspects essential to promote the practical implementation of highlighted achievements in 3PM-guided IS management. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00376-2.
Collapse
Affiliation(s)
- Ruofei Chen
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 P. R. China
| | - Xiaoyan Wang
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 P. R. China
| | - Na Li
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 P. R. China
| | - Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, University Hospital Bonn, Venusberg Campus 1, Rheinische Friedrich-Wilhelms-University of Bonn, Bonn, 53127 Germany
| | - Xianquan Zhan
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 P. R. China
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Jinan Key Laboratory of Cancer Multiomics, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Jinan, Shandong 250117 P. R. China
| |
Collapse
|
3
|
Heit BS, Chu A, McRay A, Richmond JE, Heckman CJ, Larson J. Interference with glutamate antiporter system x c - enables post-hypoxic long-term potentiation in hippocampus. Exp Physiol 2024; 109:1572-1592. [PMID: 39153228 PMCID: PMC11363115 DOI: 10.1113/ep092045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 06/28/2024] [Indexed: 08/19/2024]
Abstract
Our group previously showed that genetic or pharmacological inhibition of the cystine/glutamate antiporter, system xc -, mitigates excitotoxicity after anoxia by increasing latency to anoxic depolarization, thus attenuating the ischaemic core. Hypoxia, however, which prevails in the ischaemic penumbra, is a condition where neurotransmission is altered, but excitotoxicity is not triggered. The present study employed mild hypoxia to further probe ischaemia-induced changes in neuronal responsiveness from wild-type and xCT KO (xCT-/-) mice. Synaptic transmission was monitored in hippocampal slices from both genotypes before, during and after a hypoxic episode. Although wild-type and xCT-/- slices showed equal suppression of synaptic transmission during hypoxia, mutant slices exhibited a persistent potentiation upon re-oxygenation, an effect we termed 'post-hypoxic long-term potentiation (LTP)'. Blocking synaptic suppression during hypoxia by antagonizing adenosine A1 receptors did not preclude post-hypoxic LTP. Further examination of the induction and expression mechanisms of this plasticity revealed that post-hypoxic LTP was driven by NMDA receptor activation, as well as increased calcium influx, with no change in paired-pulse facilitation. Hence, the observed phenomenon engaged similar mechanisms as classical LTP. This was a remarkable finding as theta-burst stimulation-induced LTP was equivalent between genotypes. Importantly, post-hypoxic LTP was generated in wild-type slices pretreated with system xc - inhibitor, S-4-carboxyphenylglycine, thereby confirming the antiporter's role in this phenomenon. Collectively, these data indicate that system xc - interference enables neuroplasticity in response to mild hypoxia, and, together with its regulation of cellular damage in the ischaemic core, suggest a role for the antiporter in post-ischaemic recovery of the penumbra.
Collapse
Affiliation(s)
- Bradley S. Heit
- Department of Neuroscience and Department of Biomedical EngineeringNorthwestern UniversityChicagoIllinoisUSA
- Department of PsychiatryUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Alex Chu
- Department of PsychiatryUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Alyssa McRay
- Department of Biological SciencesUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Janet E. Richmond
- Department of Biological SciencesUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Charles J. Heckman
- Department of Neuroscience and Department of Biomedical EngineeringNorthwestern UniversityChicagoIllinoisUSA
| | - John Larson
- Department of PsychiatryUniversity of Illinois at ChicagoChicagoIllinoisUSA
- Department of Biological SciencesUniversity of Illinois at ChicagoChicagoIllinoisUSA
| |
Collapse
|
4
|
Ulger O, Eş I, Proctor CM, Algin O. Stroke studies in large animals: Prospects of mitochondrial transplantation and enhancing efficiency using hydrogels and nanoparticle-assisted delivery. Ageing Res Rev 2024; 100:102469. [PMID: 39191353 DOI: 10.1016/j.arr.2024.102469] [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: 06/12/2024] [Revised: 08/08/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
One of the most frequent reasons for mortality and disability today is acute ischemic stroke, which occurs by an abrupt disruption of cerebral circulation. The intricate damage mechanism involves several factors, such as inflammatory response, disturbance of ion balance, loss of energy production, excessive reactive oxygen species and glutamate release, and finally, neuronal death. Stroke research is now carried out using several experimental models and potential therapeutics. Furthermore, studies are being conducted to address the shortcomings of clinical care. A great deal of research is being done on novel pharmacological drugs, mitochondria targeting compounds, and different approaches including brain cooling and new technologies. Still, there are many unanswered questions about disease modeling and treatment strategies. Before these new approaches may be used in therapeutic settings, they must first be tested on large animals, as most of them have been done on rodents. However, there are several limitations to large animal stroke models used for research. In this review, the damage mechanisms in acute ischemic stroke and experimental acute ischemic stroke models are addressed. The current treatment approaches and promising experimental methods such as mitochondrial transplantation, hydrogel-based interventions, and strategies like mitochondria encapsulation and chemical modification, are also examined in this work.
Collapse
Affiliation(s)
- Oner Ulger
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, Ankara 06010, Turkiye; Gulhane Training and Research Hospital, University of Health Sciences, Ankara 06010, Turkiye.
| | - Ismail Eş
- Department of Engineering Science, Institute of Biomedical Engineering (IBME), University of Oxford, Oxford OX3 7DQ, UK
| | - Christopher M Proctor
- Department of Engineering Science, Institute of Biomedical Engineering (IBME), University of Oxford, Oxford OX3 7DQ, UK
| | - Oktay Algin
- Interventional MR Clinical R&D Institute, Ankara University, Ankara 06100, Turkiye; Department of Radiology, Medical Faculty, Ankara University, Ankara 06100, Turkiye; National MR Research Center (UMRAM), Bilkent University, Ankara 06800, Turkiye
| |
Collapse
|
5
|
Pérez-Mato M, López-Arias E, Bugallo-Casal A, Correa-Paz C, Arias S, Rodríguez-Yáñez M, Santamaría-Cadavid M, Campos F. New Perspectives in Neuroprotection for Ischemic Stroke. Neuroscience 2024; 550:30-42. [PMID: 38387732 DOI: 10.1016/j.neuroscience.2024.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/12/2024] [Accepted: 02/16/2024] [Indexed: 02/24/2024]
Abstract
The constant failure of new neuroprotective therapies for ischemic stroke has partially halted the search for new therapies in recent years, mainly because of the high investment risk required to develop a new treatment for a complex pathology, such as stroke, with a narrow intervention window and associated comorbidities. However, owing to recent progress in understanding the stroke pathophysiology, improvement in patient care in stroke units, development of new imaging techniques, search for new biomarkers for early diagnosis, and increasingly widespread use of mechanical recanalization therapies, new opportunities have opened for the study of neuroprotection. This review summarizes the main protective agents currently in use, some of which are already in the clinical evaluation phase. It also includes an analysis of how recanalization therapies, new imaging techniques, and biomarkers have improved their efficacy.
Collapse
Affiliation(s)
- María Pérez-Mato
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Esteban López-Arias
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Ana Bugallo-Casal
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Clara Correa-Paz
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Susana Arias
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, 15706 Santiago de Compostela, Spain
| | - Manuel Rodríguez-Yáñez
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, 15706 Santiago de Compostela, Spain
| | - María Santamaría-Cadavid
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, 15706 Santiago de Compostela, Spain
| | - Francisco Campos
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
6
|
Bao L, Liu Y, Jia Q, Chu S, Jiang H, He S. Argon neuroprotection in ischemic stroke and its underlying mechanism. Brain Res Bull 2024; 212:110964. [PMID: 38670471 DOI: 10.1016/j.brainresbull.2024.110964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/04/2024] [Accepted: 04/21/2024] [Indexed: 04/28/2024]
Abstract
Ischemic stroke (IS), primarily caused by cerebrovascular obstruction, results in severe neurological deficits and has emerged as a leading cause of death and disability worldwide. Recently, there has been increasing exploration of the neuroprotective properties of the inert gas argon. Argon has exhibited impressive neuroprotection in many in vivo and ex vivo experiments without signs of adverse effects, coupled with the advantages of being inexpensive and easily available. However, the efficient administration strategy and underlying mechanisms of neuroprotection by argon in IS are still unclear. This review summarizes current research on the neuroprotective effects of argon in IS with the goal to provide effective guidance for argon application and to elucidate the potential mechanisms of argon neuroprotection. Early and appropriate argon administration at as high a concentration as possible offers favorable neuroprotection in IS. Argon inhalation has been shown to provide some long-term protection benefits. Argon provides the anti-oxidative stress, anti-inflammatory and anti-apoptotic cytoprotective effects mainly around Toll-like receptor 2/4 (TLR2/4), mediated by extracellular signal-regulated kinase 1/2 (ERK1/2), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), nuclear factor kappa-B (NF-ĸB) and B-cell leukemia/lymphoma 2 (Bcl-2). Therefore, argon holds significant promise as a novel clinical neuroprotective gas agent for ischemic stroke after further researches to identify the optimal application strategy and elucidate the underlying mechanism.
Collapse
Affiliation(s)
- Li Bao
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China; Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Yongxin Liu
- Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Qi Jia
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China; Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Sihao Chu
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China; Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Han Jiang
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China; Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Shuang He
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China.
| |
Collapse
|
7
|
Barata P, Camacho O, Lima CG, Pereira AC. The Role of Hyperbaric Oxygen Therapy in Neuroregeneration and Neuroprotection: A Review. Cureus 2024; 16:e62067. [PMID: 38989389 PMCID: PMC11235151 DOI: 10.7759/cureus.62067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
Neurogenesis is a high energy-demanding process, which is why blood vessels are an active part of the neurogenic niche since they allow the much-needed oxygenation of progenitor cells. In this regard, although neglected for a long time, the "oxygen niche" should be considered an important intervenient in adult neurogenesis. One possible hypothesis for the failure of numerous neuroprotective trials is that they relied on compounds that target a highly specific neuroprotective pathway. This approach may be too limited, given the complexity of the processes that lead to cell death. Therefore, research should adopt a more multifactorial approach. Among the limited range of agents with multimodal neuromodulatory capabilities, hyperbaric oxygen therapy has demonstrated effectiveness in reducing secondary brain damage in various brain injury models. This therapy functions not only as a neuroprotective mechanism but also as a powerful neuroregenerative mechanism.
Collapse
Affiliation(s)
- Pedro Barata
- Pathology and Laboratory Medicine, Centro Hospitalar Universitário do Porto, Porto, PRT
- CECLIN (Center for Clinical Studies), Hospital-Escola da Universidade Fernando Pessoa (HE-UFP), Porto, PRT
| | - Oscar Camacho
- Hyperbaric Medicine Unit, Unidade Local de Saúde de Matosinhos, Matosinhos, PRT
| | - Clara G Lima
- Anesthesiology, Hospital Pedro Hispano, Matosinhos, PRT
| | - Ana Claudia Pereira
- Faculty of Health Sciences, Universidade Fernando Pessoa (UFP), Porto, PRT
- CECLIN (Center for Clinical Studies), Hospital-Escola da Universidade Fernando Pessoa (HE-UFP), Porto, PRT
| |
Collapse
|
8
|
Zhang J, Ren W, Liu X, Chen J, Zeng Y, Xiang H, Hu Y, Zhang H. A novel BODIPY-based theranostic agent for in vivo fluorescence imaging of cerebral Aβ and ameliorating Aβ-associated disorders in Alzheimer's disease transgenic mice. RSC Med Chem 2024; 15:1216-1224. [PMID: 38665839 PMCID: PMC11042169 DOI: 10.1039/d3md00744h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/19/2024] [Indexed: 04/28/2024] Open
Abstract
β-Amyloid (Aβ) aggregation is increasingly recognized as both a biomarker and an inducer of the progression of Alzheimer's disease (AD). Here, we describe a novel fluorescent probe P14, developed based on the BODIPY structure, capable of simultaneous visualization and inhibition of Aβ aggregation in vivo. P14 shows high binding affinity to Aβ aggregates and selectively labels Aβ plaques in the brain slices of APP/PS1 mice. Moreover, P14 is able to visualize overloaded Aβ in both APP/PS1 and 5 × FAD transgenic mice in vivo. From the aspect of potential therapeutic effects, P14 administration inhibits Aβ aggregation and alleviates Aβ-induced neuronal damage in vitro, as well as reduces central Aβ deposition and ameliorates cognitive impairment in APP/PS1 transgenic mice in vivo. Finally, P14 is applied to monitor the progression of Aβ aggregation in the brain of 5 × FAD transgenic mice and the intervention effect itself by fluorescence imaging. In summary, the discovery of this fluorescent agent might provide important clues for the future development of theranostic drug candidates targeting Aβ aggregation in AD.
Collapse
Affiliation(s)
- Jingjing Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
| | - Wenming Ren
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
| | - Xiaohui Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
| | - Jingjing Chen
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS 1 Xiangshanzhi Road Hangzhou 310024 China
| | - Yuteng Zeng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- School of Life Science and Technology, ShanghaiTech University Shanghai 201210 China
| | - Huaijiang Xiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
| | - Youhong Hu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS 1 Xiangshanzhi Road Hangzhou 310024 China
| | - Haiyan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
| |
Collapse
|
9
|
Pegoraro C, Domingo-Ortí I, Conejos-Sánchez I, Vicent MJ. Unlocking the Mitochondria for Nanomedicine-based Treatments: Overcoming Biological Barriers, Improving Designs, and Selecting Verification Techniques. Adv Drug Deliv Rev 2024; 207:115195. [PMID: 38325562 DOI: 10.1016/j.addr.2024.115195] [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: 10/24/2023] [Revised: 01/13/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
Enhanced targeting approaches will support the treatment of diseases associated with dysfunctional mitochondria, which play critical roles in energy generation and cell survival. Obstacles to mitochondria-specific targeting include the presence of distinct biological barriers and the need to pass through (or avoid) various cell internalization mechanisms. A range of studies have reported the design of mitochondrially-targeted nanomedicines that navigate the complex routes required to influence mitochondrial function; nonetheless, a significant journey lies ahead before mitochondrially-targeted nanomedicines become suitable for clinical use. Moving swiftly forward will require safety studies, in vivo assays confirming effectiveness, and methodologies to validate mitochondria-targeted nanomedicines' subcellular location/activity. From a nanomedicine standpoint, we describe the biological routes involved (from administration to arrival within the mitochondria), the features influencing rational design, and the techniques used to identify/validate successful targeting. Overall, rationally-designed mitochondria-targeted-based nanomedicines hold great promise for precise subcellular therapeutic delivery.
Collapse
Affiliation(s)
- Camilla Pegoraro
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Inés Domingo-Ortí
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Inmaculada Conejos-Sánchez
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - María J Vicent
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| |
Collapse
|
10
|
Dhawka L, Palfini V, Hambright E, Blanco I, Poon C, Kahl A, Resch U, Bhawal R, Benakis C, Balachandran V, Holder A, Zhang S, Iadecola C, Hochrainer K. Post-ischemic ubiquitination at the postsynaptic density reversibly influences the activity of ischemia-relevant kinases. Commun Biol 2024; 7:321. [PMID: 38480905 PMCID: PMC10937959 DOI: 10.1038/s42003-024-06009-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 03/04/2024] [Indexed: 03/17/2024] Open
Abstract
Ubiquitin modifications alter protein function and stability, thereby regulating cell homeostasis and viability, particularly under stress. Ischemic stroke induces protein ubiquitination at the ischemic periphery, wherein cells remain viable, however the identity of ubiquitinated proteins is unknown. Here, we employed a proteomics approach to identify these proteins in mice undergoing ischemic stroke. The data are available in a searchable web interface ( https://hochrainerlab.shinyapps.io/StrokeUbiOmics/ ). We detected increased ubiquitination of 198 proteins, many of which localize to the postsynaptic density (PSD) of glutamatergic neurons. Among these were proteins essential for maintaining PSD architecture, such as PSD95, as well as NMDA and AMPA receptor subunits. The largest enzymatic group at the PSD with elevated post-ischemic ubiquitination were kinases, such as CaMKII, PKC, Cdk5, and Pyk2, whose aberrant activities are well-known to contribute to post-ischemic neuronal death. Concurrent phospho-proteomics revealed altered PSD-associated phosphorylation patterns, indicative of modified kinase activities following stroke. PSD-located CaMKII, PKC, and Cdk5 activities were decreased while Pyk2 activity was increased after stroke. Removal of ubiquitin restored kinase activities to pre-stroke levels, identifying ubiquitination as the responsible molecular mechanism for post-ischemic kinase regulation. These findings unveil a previously unrecognized role of ubiquitination in the regulation of essential kinases involved in ischemic injury.
Collapse
Affiliation(s)
- Luvna Dhawka
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Victoria Palfini
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Emma Hambright
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Ismary Blanco
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Carrie Poon
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Anja Kahl
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Ulrike Resch
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Ruchika Bhawal
- Institute of Biotechnology, Cornell University, Ithaca, NY, USA
| | - Corinne Benakis
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Vaishali Balachandran
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Alana Holder
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Sheng Zhang
- Institute of Biotechnology, Cornell University, Ithaca, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Karin Hochrainer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
11
|
Walther J, Kirsch EM, Hellwig L, Schmerbeck SS, Holloway PM, Buchan AM, Mergenthaler P. Reinventing the Penumbra - the Emerging Clockwork of a Multi-modal Mechanistic Paradigm. Transl Stroke Res 2023; 14:643-666. [PMID: 36219377 PMCID: PMC10444697 DOI: 10.1007/s12975-022-01090-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/25/2022]
Abstract
The concept of the ischemic penumbra was originally defined as the area around a necrotic stroke core and seen as the tissue at imminent risk of further damage. Today, the penumbra is generally considered as time-sensitive hypoperfused brain tissue with decreased oxygen and glucose availability, salvageable tissue as treated by intervention, and the potential target for neuroprotection in focal stroke. The original concept entailed electrical failure and potassium release but one short of neuronal cell death and was based on experimental stroke models, later confirmed in clinical imaging studies. However, even though the basic mechanisms have translated well, conferring brain protection, and improving neurological outcome after stroke based on the pathophysiological mechanisms in the penumbra has yet to be achieved. Recent findings shape the modern understanding of the penumbra revealing a plethora of molecular and cellular pathophysiological mechanisms. We now propose a new model of the penumbra, one which we hope will lay the foundation for future translational success. We focus on the availability of glucose, the brain's central source of energy, and bioenergetic failure as core pathophysiological concepts. We discuss the relation of mitochondrial function in different cell types to bioenergetics and apoptotic cell death mechanisms, autophagy, and neuroinflammation, to glucose metabolism in what is a dynamic ischemic penumbra.
Collapse
Affiliation(s)
- Jakob Walther
- Charité - Universitätsmedizin Berlin, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Elena Marie Kirsch
- Charité - Universitätsmedizin Berlin, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Lina Hellwig
- Charité - Universitätsmedizin Berlin, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Sarah S Schmerbeck
- Charité - Universitätsmedizin Berlin, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Paul M Holloway
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK
| | - Alastair M Buchan
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK.
| | - Philipp Mergenthaler
- Charité - Universitätsmedizin Berlin, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, NeuroCure Clinical Research Center, Charitéplatz 1, 10117, Berlin, Germany.
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK.
| |
Collapse
|
12
|
Dhawka L, Palfini V, Hambright E, Blanco I, Poon C, Kahl A, Resch U, Bhawal R, Benakis C, Balachandran V, Zhang S, Iadecola C, Hochrainer K. Post-ischemic ubiquitination at the postsynaptic density reversibly influences the activity of ischemia-relevant kinases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.21.552860. [PMID: 37662420 PMCID: PMC10473581 DOI: 10.1101/2023.08.21.552860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Ubiquitin modifications alter protein function and stability, thereby regulating cell homeostasis and viability, particularly under stress. Ischemic stroke induces protein ubiquitination at the ischemic periphery, wherein cells remain viable, however the identity of ubiquitinated proteins is unknown. Here, we employed a proteomics approach to identify these proteins in mice undergoing ischemic stroke. The data are available in a searchable web interface ( https://hochrainerlab.shinyapps.io/StrokeUbiOmics/ ). We detected increased ubiquitination of 198 proteins, many of which localize to the postsynaptic density (PSD) of glutamatergic neurons. Among these were proteins essential for maintaining PSD architecture, such as PSD95, as well as NMDA and AMPA receptor subunits. The largest enzymatic group at the PSD with elevated post-ischemic ubiquitination were kinases, such as CaMKII, PKC, Cdk5, and Pyk2, whose aberrant activities are well-known to contribute to post-ischemic neuronal death. Concurrent phospho-proteomics revealed altered PSD-associated phosphorylation patterns, indicative of modified kinase activities following stroke. PSD-located CaMKII, PKC, and Cdk5 activities were decreased while Pyk2 activity was increased after stroke. Removal of ubiquitin restored kinase activities to pre-stroke levels, identifying ubiquitination as the responsible molecular mechanism for post-ischemic kinase regulation. These findings unveil a previously unrecognized role of ubiquitination in the regulation of essential kinases involved in ischemic injury.
Collapse
|
13
|
Okonkwo ON, Agweye CT, Akanbi T. Neuroprotection for Nonarteritic Central Retinal Artery Occlusion: Lessons from Acute Ischemic Stroke. Clin Ophthalmol 2023; 17:1531-1543. [PMID: 37284058 PMCID: PMC10239763 DOI: 10.2147/opth.s403433] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/19/2023] [Indexed: 06/08/2023] Open
Abstract
Nonarteritic central retinal artery occlusion (NA-CRAO) is a variant of acute ischemic stroke (AIS) and is a cause of sudden severe loss of vision. There are guidelines by the American Heart Association and the American Stroke Association for the care of CRAO patients. This review explores the basis of retinal neuroprotection for CRAO and its potential for improving the outcome of NA-CRAO. Recently, there have been significant advances in research into the use of neuroprotection to treat retinal diseases, including retinal detachment, age-related macular degeneration, and inherited retinal diseases. Also, neuroprotective research in AIS has been extensive, and newer drugs tested, including Uric acid, Nerinetide, and Otaplimastat, with promising results. Progress in cerebral neuroprotection after AIS offers hope for retinal neuroprotection after CRAO; and a possibility of extrapolating research findings from AIS into CRAO. Combining neuroprotection and thrombolysis can extend the therapeutic window for NA-CRAO treatment and potentially improve outcomes. Experimented neuroprotection for CRAO includes Angiopoietin (Comp Ang1), KUS 121, Gene therapy (XIAP), and hypothermia. Efforts in the field of neuroprotection for NA-CRAO should focus on better imaging to delineate the penumbra after an acute episode of NA-CRAO (using a combination of high-definition optical coherence angiography and electrophysiology). Also, research should explore details of pathophysiologic mechanisms involved in NA-CRAO, allowing for further neuroprotective intervention, and closing the gap between preclinical and clinical neuroprotection.
Collapse
Affiliation(s)
- Ogugua Ndubuisi Okonkwo
- Department of Ophthalmology, Eye Foundation Hospital and Eye Foundation Retina Institute, Ikeja, Lagos, Nigeria
| | - Chineze Thelma Agweye
- Department of Ophthalmology, University of Calabar and University of Calabar Teaching Hospital, Cross River, Nigeria
| | - Toyin Akanbi
- Department of Ophthalmology, Eye Foundation Hospital and Eye Foundation Retina Institute, Ikeja, Lagos, Nigeria
| |
Collapse
|
14
|
Li J, Dai F, Kou X, Wu B, Xu J, He S. β-Actin: An Emerging Biomarker in Ischemic Stroke. Cell Mol Neurobiol 2023; 43:683-696. [PMID: 35556192 PMCID: PMC11415192 DOI: 10.1007/s10571-022-01225-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/10/2022] [Indexed: 11/03/2022]
Abstract
At present, the diagnosis of ischemic stroke mainly depends on neuroimaging technology, but it still has many limitations. Therefore, it is very important to find new biomarkers of ischemic stroke. Recently, β-actin has attracted extensive attention as a biomarker of a variety of cancers. Although several recent studies have been investigating its role in ischemic stroke and other cerebrovascular diseases, the understanding of this emerging biomarker in neurology is still limited. We examined human and preclinical studies to gain a comprehensive understanding of the literature on the subject. Most relevant literatures focus on preclinical research, and pay more attention to the role of β-actin in the process of cerebral ischemia, but some recent literatures reported that in human studies, serum β-actin increased significantly in the early stage of acute cerebral ischemia. This review will investigate the basic biology of β-actin, pay attention to the potential role of serum β-actin as an early diagnostic blood biomarker of ischemic stroke, and explore its potential mechanism in ischemic stroke and new strategies for stroke treatment in the future.
Collapse
Affiliation(s)
- Jiaqian Li
- Department of Neurology, School of Medicine, Zhoushan Hospital, Zhejiang University, Zhoushan, 316000, Zhejiang Province, China
| | - Fangyu Dai
- Department of Neurology, School of Medicine, Zhoushan Hospital, Zhejiang University, Zhoushan, 316000, Zhejiang Province, China
| | - Xuelian Kou
- Department of Neurology, School of Medicine, Zhoushan Hospital, Zhejiang University, Zhoushan, 316000, Zhejiang Province, China
| | - Bin Wu
- Department of Neurology, School of Medicine, Zhoushan Hospital, Zhejiang University, Zhoushan, 316000, Zhejiang Province, China
| | - Jie Xu
- Department of Neurology, School of Medicine, Zhoushan Hospital, Zhejiang University, Zhoushan, 316000, Zhejiang Province, China
| | - Songbin He
- Department of Neurology, School of Medicine, Zhoushan Hospital, Zhejiang University, Zhoushan, 316000, Zhejiang Province, China.
| |
Collapse
|
15
|
Morris GP, Gowing EK, Courtney J, Coombe HE, King NE, Rewell SSJ, Howells DW, Clarkson AN, Sutherland BA. Vascular perfusion differs in two distinct PDGFRβ-positive zones within the ischemic core of male mice 2 weeks following photothrombotic stroke. J Neurosci Res 2023; 101:278-292. [PMID: 36412274 PMCID: PMC10952185 DOI: 10.1002/jnr.25146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 10/07/2022] [Accepted: 11/06/2022] [Indexed: 11/23/2022]
Abstract
Stroke therapy has largely focused on preventing damage and encouraging repair outside the ischemic core, as the core is considered irreparable. Recently, several studies have suggested endogenous responses within the core are important for limiting the spread of damage and enhancing recovery, but the role of blood flow and capillary pericytes in this process is unknown. Using the Rose Bengal photothrombotic model of stroke, we illustrate blood vessels are present in the ischemic core and peri-lesional regions 2 weeks post stroke in male mice. A FITC-albumin gel cast of the vasculature revealed perfusion of these vessels, suggesting cerebral blood flow (CBF) may be partially present, without vascular leakage. The length of these vessels is significantly reduced compared to uninjured regions, but the average width is greater, suggesting they are either larger vessels that survived the initial injury, smaller vessels that have expanded in size (i.e., arteriogenesis), or that neovascularization begins with larger vessels. Concurrently, we observed an increase in platelet-derived growth factor receptor beta (PDGFRβ, a marker of pericytes) expression within the ischemic core in two distinct patterns, one which resembles pericyte-derived fibrotic scarring at the edge of the core, and one which is vessel associated and may represent blood vessel recovery. We find little evidence for dividing cells on these intralesional blood vessels 2 weeks post stroke. Our study provides evidence flow is present in PDGFRβ-positive vessels in the ischemic core 2 weeks post stroke. We hypothesize intralesional CBF is important for limiting injury and for encouraging endogenous repair following cerebral ischemia.
Collapse
Affiliation(s)
- Gary P. Morris
- Tasmanian School of Medicine, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Emma K. Gowing
- Department of Anatomy, Brain Health Research Centre and Brain Research New ZealandUniversity of OtagoDunedinNew Zealand
| | - Jo‐Maree Courtney
- Tasmanian School of Medicine, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Hannah E. Coombe
- Tasmanian School of Medicine, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Natalie E. King
- Tasmanian School of Medicine, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Sarah S. J. Rewell
- Florey Institute of Neuroscience and Mental HealthMelbourne Brain Centre, Austin CampusHeidelbergVictoriaAustralia
| | - David W. Howells
- Tasmanian School of Medicine, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Andrew N. Clarkson
- Department of Anatomy, Brain Health Research Centre and Brain Research New ZealandUniversity of OtagoDunedinNew Zealand
| | - Brad A. Sutherland
- Tasmanian School of Medicine, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| |
Collapse
|
16
|
McClendon LK, Garcia RL, Lazaro T, Robledo A, Vasandani V, Luna ZAE, Rao AS, Srivatsan A, Lonard DM, Dacso CC, Kan P, O’Malley BW. A steroid receptor coactivator small molecule "stimulator" attenuates post-stroke ischemic brain injury. Front Mol Neurosci 2022; 15:1055295. [PMID: 36533127 PMCID: PMC9751323 DOI: 10.3389/fnmol.2022.1055295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/10/2022] [Indexed: 11/15/2023] Open
Abstract
Introduction: Pathologic remodeling of the brain following ischemic stroke results in neuronal loss, increased inflammation, oxidative stress, astrogliosis, and a progressive decrease in brain function. We recently demonstrated that stimulation of steroid receptor coactivator 3 with the small-molecule stimulator MCB-613 improves cardiac function in a mouse model of myocardial ischemia. Since steroid receptor coactivators are ubiquitously expressed in the brain, we reasoned that an MCB-613 derivative (MCB-10-1), could protect the brain following ischemic injury. To test this, we administered MCB-10-1 to rats following middle cerebral artery occlusion and reperfusion. Methods: Neurologic impairment and tissue damage responses were evaluated on day 1 and day 4 following injury in rats treated with control or 10-1. Results: We show that 10-1 attenuates injury post-stroke. 10-1 decreases infarct size and mitigates neurologic impairment. When given within 30 min post middle cerebral artery occlusion and reperfusion, 10-1 induces lasting protection from tissue damage in the ischemic penumbra concomitant with: (1) promotion of reparative microglia; (2) an increase in astrocyte NRF2 and GLT-1 expression; (3) early microglia activation; and (4) attenuation of astrogliosis. Discussion: Steroid receptor coactivator stimulation with MCB-10-1 is a potential therapeutic strategy for reducing inflammation and oxidative damage that cause neurologic impairment following an acute ischemic stroke.
Collapse
Affiliation(s)
- Lisa K. McClendon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- CoRegen, Inc., Baylor College of Medicine, Houston, TX, United States
| | - Roberto L. Garcia
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX, United States
| | - Tyler Lazaro
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, United States
| | - Ariadna Robledo
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX, United States
| | - Viren Vasandani
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX, United States
| | - Zean Aaron Evan Luna
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX, United States
| | - Abhijit S. Rao
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX, United States
| | - Aditya Srivatsan
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, United States
| | - David M. Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- CoRegen, Inc., Baylor College of Medicine, Houston, TX, United States
| | - Clifford C. Dacso
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- CoRegen, Inc., Baylor College of Medicine, Houston, TX, United States
| | - Peter Kan
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX, United States
| | - Bert W. O’Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- CoRegen, Inc., Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
17
|
He Y, Ying J, Tang J, Zhou R, Qu H, Qu Y, Mu D. Neonatal Arterial Ischaemic Stroke: Advances in Pathologic Neural Death, Diagnosis, Treatment, and Prognosis. Curr Neuropharmacol 2022; 20:2248-2266. [PMID: 35193484 PMCID: PMC9890291 DOI: 10.2174/1570159x20666220222144744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/04/2022] [Accepted: 02/18/2022] [Indexed: 12/29/2022] Open
Abstract
Neonatal arterial ischaemic stroke (NAIS) is caused by focal arterial occlusion and often leads to severe neurological sequelae. Neural deaths after NAIS mainly include necrosis, apoptosis, necroptosis, autophagy, ferroptosis, and pyroptosis. These neural deaths are mainly caused by upstream stimulations, including excitotoxicity, oxidative stress, inflammation, and death receptor pathways. The current clinical approaches to managing NAIS mainly focus on supportive treatments, including seizure control and anticoagulation. In recent years, research on the pathology, early diagnosis, and potential therapeutic targets of NAIS has progressed. In this review, we summarise the latest progress of research on the pathology, diagnosis, treatment, and prognosis of NAIS and highlight newly potential diagnostic and treatment approaches.
Collapse
Affiliation(s)
- Yang He
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Junjie Ying
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Jun Tang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Ruixi Zhou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Haibo Qu
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| |
Collapse
|
18
|
Safiullov Z, Izmailov A, Sokolov M, Markosyan V, Kundakchan G, Garifulin R, Shmarov M, Naroditsky B, Logunov D, Islamov R. Autologous Genetically Enriched Leucoconcentrate in the Preventive and Acute Phases of Stroke Treatment in a Mini-Pig Model. Pharmaceutics 2022; 14:pharmaceutics14102209. [PMID: 36297644 PMCID: PMC9611398 DOI: 10.3390/pharmaceutics14102209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/21/2022] [Accepted: 10/12/2022] [Indexed: 12/05/2022] Open
Abstract
The natural limitations of regeneration in the CNS are major problems for the treatment of neurological disorders, including ischaemic brain strokes. Among the approaches being actively developed to inhibit post-ischaemic negative consequences is the delivery of therapeutic genes encoding neuroprotective molecules to the brain. Unfortunately, there are currently no proven and available medicines that contain recombinant human genes for the treatment of ischaemic cerebral stroke. Of particular interest is the development of treatments for patients at risk of ischaemic stroke. In the present study, we propose a proof of concept for the use of an autologous, genetically enriched leucoconcentrate temporally secreting recombinant vascular endothelial growth factor (VEGF), glial-cell-line-derived neurotrophic factor (GDNF) and the neural cell adhesion molecule (NCAM) for the treatment of stroke. In a mini-pig ischaemic stroke model, genetically enriched leucoconcentrate was infused 4 h after surgery (gene therapy in acute phase) or 2 days before stroke modelling (preventive gene therapy). On day 21, after the stroke modelling, the post-ischaemic brain recovery was examined by morphologic and immunofluorescence analysis. The benefits of treating a stroke with genetically enriched leucoconcentrate both for preventive purposes and in the acute phase were confirmed by an improved performance in behavioural tests, higher preservation of brain tissue and positive post-ischaemic brain remodelling in the peri-infarct area. These results suggest that the employment of autologous leucocytes enabling the temporary production of the recombinant therapeutic molecules to correct the pathological process in the CNS may be one of the breakthrough approaches in gene therapy.
Collapse
Affiliation(s)
- Zufar Safiullov
- The Department of Histology, Cytology and Embryology, Kazan State Medical University, 420012 Kazan, Russia
| | - Andrei Izmailov
- The Department of Histology, Cytology and Embryology, Kazan State Medical University, 420012 Kazan, Russia
| | - Mikhail Sokolov
- The Department of Histology, Cytology and Embryology, Kazan State Medical University, 420012 Kazan, Russia
| | - Vage Markosyan
- The Department of Histology, Cytology and Embryology, Kazan State Medical University, 420012 Kazan, Russia
| | - Grayr Kundakchan
- The Department of Histology, Cytology and Embryology, Kazan State Medical University, 420012 Kazan, Russia
| | - Ravil Garifulin
- The Department of Histology, Cytology and Embryology, Kazan State Medical University, 420012 Kazan, Russia
| | - Maksim Shmarov
- The National Research Center for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Boris Naroditsky
- The National Research Center for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Denis Logunov
- The National Research Center for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Rustem Islamov
- The Department of Histology, Cytology and Embryology, Kazan State Medical University, 420012 Kazan, Russia
- Correspondence:
| |
Collapse
|
19
|
Custodia A, Ouro A, Sargento-Freitas J, Aramburu-Núñez M, Pías-Peleteiro JM, Hervella P, Rosell A, Ferreira L, Castillo J, Romaus-Sanjurjo D, Sobrino T. Unraveling the potential of endothelial progenitor cells as a treatment following ischemic stroke. Front Neurol 2022; 13:940682. [PMID: 36158970 PMCID: PMC9492921 DOI: 10.3389/fneur.2022.940682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Ischemic stroke is becoming one of the most common causes of death and disability in developed countries. Since current therapeutic options are quite limited, focused on acute reperfusion therapies that are hampered by a very narrow therapeutic time window, it is essential to discover novel treatments that not only stop the progression of the ischemic cascade during the acute phase, but also improve the recovery of stroke patients during the sub-acute or chronic phase. In this regard, several studies have shown that endothelial progenitor cells (EPCs) can repair damaged vessels as well as generate new ones following cerebrovascular damage. EPCs are circulating cells with characteristics of both endothelial cells and adult stem cells presenting the ability to differentiate into mature endothelial cells and self-renew, respectively. Moreover, EPCs have the advantage of being already present in healthy conditions as circulating cells that participate in the maintenance of the endothelium in a direct and paracrine way. In this scenario, EPCs appear as a promising target to tackle stroke by self-promoting re-endothelization, angiogenesis and vasculogenesis. Based on clinical data showing a better neurological and functional outcome in ischemic stroke patients with higher levels of circulating EPCs, novel and promising therapeutic approaches would be pharmacological treatment promoting EPCs-generation as well as EPCs-based therapies. Here, we will review the latest advances in preclinical as well as clinical research on EPCs application following stroke, not only as a single treatment but also in combination with new therapeutic approaches.
Collapse
Affiliation(s)
- Antía Custodia
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Alberto Ouro
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - João Sargento-Freitas
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
- Centro Neurociências e Biologia Celular, Coimbra, Portugal
| | - Marta Aramburu-Núñez
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Juan Manuel Pías-Peleteiro
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Pablo Hervella
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Lino Ferreira
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
- Centro Neurociências e Biologia Celular, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, UC, Biotech Parque Tecnológico de Cantanhede, University of Coimbra, Coimbra, Portugal
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Daniel Romaus-Sanjurjo
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- *Correspondence: Daniel Romaus-Sanjurjo
| | - Tomás Sobrino
- NeuroAging Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Tomás Sobrino
| |
Collapse
|
20
|
Zhou X, Nan Y, Ju J, Zhou J, Xiao H, Wang S. Comparison of Two Software Packages for Perfusion Imaging: Ischemic Core and Penumbra Estimation and Patient Triage in Acute Ischemic Stroke. Cells 2022; 11:2547. [PMID: 36010624 PMCID: PMC9406974 DOI: 10.3390/cells11162547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/20/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
Purpose: Automated postprocessing packages have been developed for managing acute ischemic stroke (AIS). These packages identify ischemic core and penumbra using either computed tomographic perfusion imaging (CTP) data or magnetic resonance imaging (MRI) data. Measurements of abnormal tissues and treatment decisions derived from different vendors can vary. The purpose of this study is to investigate the agreement of volumetric and decision-making outcomes derived from two software packages. Methods: A total of 594 AIS patients (174 underwent CTP and 420 underwent MRI) were included. Imaging data were accordingly postprocessed by two software packages: RAPID and RealNow. Volumetric outputs were compared between packages by performing intraclass correlation coefficient (ICC), Wilcoxon paired test and Bland-Altman analysis. Concordance of selecting patients eligible for mechanical thrombectomy (MT) was assessed based on neuroimaging criteria proposed in DEFUSE3. Results: In the group with CTP data, mean ischemic core volume (ICV)/penumbral volume (PV) was 14.9/81.1 mL via RAPID and 12.6/83.2 mL via RealNow. Meanwhile, in the MRI group, mean ICV/PV were 52.4/68.4 mL and 48.9/61.6 mL via RAPID and RealNow, respectively. Reliability, which was measured by ICC of ICV and PV in CTP and MRI groups, ranged from 0.87 to 0.99. The bias remained small between measurements (CTP ICV: 0.89 mL, CTP PV: -2 mL, MRI ICV: 3.5 mL and MRI PV: 6.8 mL). In comparison with CTP ICV with follow-up DWI, the ICC was 0.92 and 0.94 for RAPID and Realnow, respectively. The bias remained small between CTP ICV and follow-up DWI measurements (Rapid: -4.65 mL, RealNow: -3.65 mL). Wilcoxon paired test showed no significant difference between measurements. The results of patient triage were concordant in 159/174 cases (91%, ICC: 0.90) for CTP and 400/420 cases (95%, ICC: 0.93) for MRI. Conclusion: The CTP ICV derived from RealNow was more accurate than RAPID. The similarity in volumetric measurement between packages did not necessarily relate to equivalent patient triage. In this study, RealNow showed excellent agreement with RAPID in measuring ICV and PV as well as patient triage.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Rd., Shanghai 200065, China
| | - Yashi Nan
- YIWEI Medical Technology Co., Ltd., Room 1001, MAI KE LONG Building, Shenzhen 518000, China
| | - Jieyang Ju
- The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan Rd., Nanjing 210011, China
| | - Jingyu Zhou
- YIWEI Medical Technology Co., Ltd., Room 1001, MAI KE LONG Building, Shenzhen 518000, China
| | - Huanhui Xiao
- YIWEI Medical Technology Co., Ltd., Room 1001, MAI KE LONG Building, Shenzhen 518000, China
| | - Silun Wang
- YIWEI Medical Technology Co., Ltd., Room 1001, MAI KE LONG Building, Shenzhen 518000, China
| |
Collapse
|
21
|
Metabolomics-Based Pharmacodynamic Analysis of Zhuang Yao Shuang Lu Tong Nao Granules in a Rat Model of Ischemic Cerebral Infarction. Anal Cell Pathol (Amst) 2022; 2022:8776079. [PMID: 35846873 PMCID: PMC9277214 DOI: 10.1155/2022/8776079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/30/2022] [Accepted: 06/04/2022] [Indexed: 12/04/2022] Open
Abstract
This study used a metabolomic approach to reveal changes in the levels of metabolic biomarkers and related metabolic pathways before and after Zhuang Yao Shuang Lu Tong Nao granule (YHT) treatment in rats with cerebral ischemia. The neurological deficit scores were significantly higher in the MCAO_R group than in the NC group, indicating that the mice had significantly impaired motor functions. The YHT group had significantly lower scores than the MCAO_R group, suggesting that YHT significantly improved motor function in rats. TTC staining of the brain tissue revealed that YHT significantly reduced the area of cerebral infarction in the treated rats. The MCAO_R group was better separated from the NC rent, sham, and YHT groups via metabolomic PCA. Moreover, there were significant differences in the differential metabolites between the MACO_R and YHT groups. Eighteen common differential metabolites were detected between the MACO_R and NC groups, MACO_R and sham groups, and MACO_R and YHT groups, indicating that YHT significantly increased the levels of various metabolites in the serum of cerebral ischemic stroke (CIS) rats. Moreover, a total of 23 metabolic pathways were obtained. We identified 11 metabolic pathways with the most significant effects in the bubble plots. In conclusion, from a systems biology perspective, this metabolomics-based study showed that YHT could be used to treat ischemic stroke by modulating changes in endogenous metabolites.
Collapse
|
22
|
Polyphenols for the Treatment of Ischemic Stroke: New Applications and Insights. Molecules 2022; 27:molecules27134181. [PMID: 35807426 PMCID: PMC9268254 DOI: 10.3390/molecules27134181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
Ischemic stroke (IS) is a leading cause of death and disability worldwide. Currently, the main therapeutic strategy involves the use of intravenous thrombolysis to restore cerebral blood flow to prevent the transition of the penumbra to the infarct core. However, due to various limitations and complications, including the narrow time window in which this approach is effective, less than 10% of patients benefit from such therapy. Thus, there is an urgent need for alternative therapeutic strategies, with neuroprotection against the ischemic cascade response after IS being one of the most promising options. In the past few decades, polyphenolic compounds have shown great potential in animal models of IS because of their high biocompatibility and ability to target multiple ischemic cascade signaling pathways, although low bioavailability is an issue that limits the applications of several polyphenols. Here, we review the pathophysiological changes following cerebral ischemia and summarize the research progress regarding the applications of polyphenolic compounds in the treatment of IS over the past 5 years. Furthermore, we discuss several potential strategies for improving the bioavailability of polyphenolic compounds as well as some essential issues that remain to be addressed for the translation of the related therapies to the clinic.
Collapse
|
23
|
Ghozy S, Reda A, Varney J, Elhawary AS, Shah J, Murry K, Sobeeh MG, Nayak SS, Azzam AY, Brinjikji W, Kadirvel R, Kallmes DF. Neuroprotection in Acute Ischemic Stroke: A Battle Against the Biology of Nature. Front Neurol 2022; 13:870141. [PMID: 35711268 PMCID: PMC9195142 DOI: 10.3389/fneur.2022.870141] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/21/2022] [Indexed: 12/22/2022] Open
Abstract
Stroke is the second most common cause of global death following coronary artery disease. Time is crucial in managing stroke to reduce the rapidly progressing insult of the ischemic penumbra and the serious neurologic deficits that might follow it. Strokes are mainly either hemorrhagic or ischemic, with ischemic being the most common of all types of strokes. Thrombolytic therapy with recombinant tissue plasminogen activator and endovascular thrombectomy are the main types of management of acute ischemic stroke (AIS). In addition, there is a vital need for neuroprotection in the setting of AIS. Neuroprotective agents are important to investigate as they may reduce mortality, lessen disability, and improve quality of life after AIS. In our review, we will discuss the main types of management and the different modalities of neuroprotection, their mechanisms of action, and evidence of their effectiveness after ischemic stroke.
Collapse
Affiliation(s)
- Sherief Ghozy
- Department of Neuroradiology, Mayo Clinic, Rochester, MN, United States.,Nuffield Department of Primary Care Health Sciences and Department for Continuing Education (EBHC Program), Oxford University, Oxford, United Kingdom
| | - Abdullah Reda
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Joseph Varney
- School of Medicine, American University of the Caribbean, Philipsburg, Sint Maarten
| | | | - Jaffer Shah
- Medical Research Center, Kateb University, Kabul, Afghanistan
| | | | - Mohamed Gomaa Sobeeh
- Faculty of Physical Therapy, Sinai University, Cairo, Egypt.,Faculty of Physical Therapy, Cairo University, Giza, Egypt
| | - Sandeep S Nayak
- Department of Internal Medicine, NYC Health + Hospitals/Metropolitan, New York, NY, United States
| | - Ahmed Y Azzam
- Faculty of Medicine, October 6 University, Giza, Egypt
| | - Waleed Brinjikji
- Department of Neurosurgery, Mayo Clinic Rochester, Rochester, MN, United States
| | | | - David F Kallmes
- Department of Neuroradiology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
24
|
Ouro A, Correa-Paz C, Maqueda E, Custodia A, Aramburu-Núñez M, Romaus-Sanjurjo D, Posado-Fernández A, Candamo-Lourido M, Alonso-Alonso ML, Hervella P, Iglesias-Rey R, Castillo J, Campos F, Sobrino T. Involvement of Ceramide Metabolism in Cerebral Ischemia. Front Mol Biosci 2022; 9:864618. [PMID: 35531465 PMCID: PMC9067562 DOI: 10.3389/fmolb.2022.864618] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/11/2022] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke, caused by the interruption of blood flow to the brain and subsequent neuronal death, represents one of the main causes of disability in worldwide. Although reperfusion therapies have shown efficacy in a limited number of patients with acute ischemic stroke, neuroprotective drugs and recovery strategies have been widely assessed, but none of them have been successful in clinical practice. Therefore, the search for new therapeutic approaches is still necessary. Sphingolipids consist of a family of lipidic molecules with both structural and cell signaling functions. Regulation of sphingolipid metabolism is crucial for cell fate and homeostasis in the body. Different works have emphasized the implication of its metabolism in different pathologies, such as diabetes, cancer, neurodegeneration, or atherosclerosis. Other studies have shown its implication in the risk of suffering a stroke and its progression. This review will highlight the implications of sphingolipid metabolism enzymes in acute ischemic stroke.
Collapse
Affiliation(s)
- Alberto Ouro
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Clara Correa-Paz
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Elena Maqueda
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Antía Custodia
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Marta Aramburu-Núñez
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Daniel Romaus-Sanjurjo
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Adrián Posado-Fernández
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - María Candamo-Lourido
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Maria Luz Alonso-Alonso
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Pablo Hervella
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Ramón Iglesias-Rey
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Francisco Campos
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Tomás Sobrino
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| |
Collapse
|
25
|
Wang L, Ren W, Wu Q, Liu T, Wei Y, Ding J, Zhou C, Xu H, Yang S. NLRP3 Inflammasome Activation: A Therapeutic Target for Cerebral Ischemia–Reperfusion Injury. Front Mol Neurosci 2022; 15:847440. [PMID: 35600078 PMCID: PMC9122020 DOI: 10.3389/fnmol.2022.847440] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 04/06/2022] [Indexed: 12/16/2022] Open
Abstract
Millions of patients are suffering from ischemic stroke, it is urgent to figure out the pathogenesis of cerebral ischemia–reperfusion (I/R) injury in order to find an effective cure. After I/R injury, pro-inflammatory cytokines especially interleukin-1β (IL-1β) upregulates in ischemic brain cells, such as microglia and neuron. To ameliorate the inflammation after cerebral I/R injury, nucleotide-binding oligomerization domain (NOD), leucine-rich repeat (LRR), and pyrin domain-containing protein 3 (NLRP3) inflammasome is well-investigated. NLRP3 inflammasomes are complicated protein complexes that are activated by endogenous and exogenous danger signals to participate in the inflammatory response. The assembly and activation of the NLRP3 inflammasome lead to the caspase-1-dependent release of pro-inflammatory cytokines, such as interleukin (IL)-1β and IL-18. Furthermore, pyroptosis is a pro-inflammatory cell death that occurs in a dependent manner on NLRP3 inflammasomes after cerebral I/R injury. In this review, we summarized the assembly and activation of NLRP3 inflammasome; moreover, we also concluded the pivotal role of NLRP3 inflammasome and inhibitors, targeting the NLRP3 inflammasome in cerebral I/R injury.
Collapse
Affiliation(s)
- Lixia Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Ren
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Qingjuan Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tianzhu Liu
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Ying Wei
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiru Ding
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chen Zhou
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Houping Xu
- Preventive Treatment Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Houping Xu
| | - Sijin Yang
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Sijin Yang
| |
Collapse
|
26
|
CD40-CD40L in Neurological Disease. Int J Mol Sci 2022; 23:ijms23084115. [PMID: 35456932 PMCID: PMC9031401 DOI: 10.3390/ijms23084115] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/02/2022] [Accepted: 04/04/2022] [Indexed: 02/06/2023] Open
Abstract
Immune-inflammatory conditions in the central nervous system (CNS) rely on molecular and cellular interactions which are homeostatically maintained to protect neural tissue from harm. The CD40–CD40L interaction upregulates key proinflammatory molecules, a function best understood in the context of infection, during which B-cells are activated via CD40 signaling to produce antibodies. However, the role of CD40 in neurological disease of non-infectious etiology is unclear. We review the role of CD40–CD40L in traumatic brain injury, Alzheimer’s Disease, Parkinson’s Disease, stroke, epilepsy, nerve injury, multiple sclerosis, ALS, myasthenia gravis and brain tumors. We also highlight therapeutic advancements targeting the CD40 system to either attenuate the neuroinflammatory response or leverage the downstream effects of CD40 signaling for direct tumor cell lysis.
Collapse
|
27
|
Han Q, Yang J, Gao X, Li J, Wu Y, Xu Y, Shang Q, Parsons MW, Lin L. Early Edema Within the Ischemic Core Is Time-Dependent and Associated With Functional Outcomes of Acute Ischemic Stroke Patients. Front Neurol 2022; 13:861289. [PMID: 35463133 PMCID: PMC9021998 DOI: 10.3389/fneur.2022.861289] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/08/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveTo investigate the difference in early edema, quantified by net water uptake (NWU) based on computed tomography (CT) between ischemic core and penumbra and to explore predictors of NWU and test its predictive power for clinical outcome.MethodsRetrospective analysis was conducted on patients admitted to Ningbo First Hospital with anterior circulation stroke and multi-modal CT. In 154 included patients, NWU of the ischemic core and penumbra were calculated and compared by Mann–Whitney U test. Correlations between NWU and variables including age, infarct time (time from symptom onset to imaging), volume of ischemic core, collateral status, and National Institutes of Health Stroke Scale (NIHSS) scores were investigated by Spearman's correlation analyses. Clinical outcome was defined using the modified Rankin Scale (mRS) at 90 days. Logistic regression and receiver operating characteristic analyses were performed to test the predictive value of NWU. Summary statistics are presented as median (interquartile range), mean (standard deviation) or estimates (95% confidence interval).ResultsThe NWU within the ischemic core [6.1% (2.9–9.2%)] was significantly higher than that of the penumbra [1.8% (−0.8–4.0%)]. The only significant predictor of NWU within the ischemic core was infarct time (p = 0.004). The NWU within the ischemic core [odds ratio = 1.23 (1.10–1.39)], the volume of ischemic core [1.04, (1.02–1.06)], age [1.09 (1.01–1.17)], and admission NHISS score [1.05 (1.01–1.09)] were associated with the outcome of patients adjusted for sex and treatment. The predictive power for the outcome of the model was significantly higher when NWU was included (area under the curve 0.875 vs. 0.813, p < 0.05 by Delong test).ConclusionsEarly edema quantified by NWU is relatively limited in the ischemic core and develops in a time-dependent manner. NWU estimates within the ischemic core may help to predict clinical outcomes of patients with acute ischemic stroke.
Collapse
Affiliation(s)
- Qing Han
- Department of Neurology, Ningbo First Hospital, Ningbo, China
| | - Jianhong Yang
- Department of Neurology, Ningbo First Hospital, Ningbo, China
| | - Xiang Gao
- Department of Neurosurgery, Ningbo First Hospital, Ningbo, China
| | - Jichuan Li
- Department of Neurology, Ningbo First Hospital, Ningbo, China
| | - Yuefei Wu
- Department of Neurology, Ningbo First Hospital, Ningbo, China
| | - Yao Xu
- Department of Neurology, Ningbo First Hospital, Ningbo, China
| | - Qing Shang
- Department of Neurology, Ningbo First Hospital, Ningbo, China
| | - Mark W. Parsons
- Sydney Brain Center, University of New South Wales, Sydney, NSW, Australia
- Mark W. Parsons
| | - Longting Lin
- Department of Neurology, Ningbo First Hospital, Ningbo, China
- Sydney Brain Center, University of New South Wales, Sydney, NSW, Australia
- *Correspondence: Longting Lin
| |
Collapse
|
28
|
Stress Granules and Acute Ischemic Stroke: Beyond mRNA Translation. Int J Mol Sci 2022; 23:ijms23073747. [PMID: 35409112 PMCID: PMC8998762 DOI: 10.3390/ijms23073747] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/25/2022] [Accepted: 03/26/2022] [Indexed: 02/06/2023] Open
Abstract
Ischemic stroke is a leading cause of death and disability worldwide. Following an ischemic insult, cells undergo endoplasmic reticulum (ER) stress, which increases the ER’s protein-folding and degradative capacities and blocks the global synthesis of proteins by phosphorylating the eukaryotic translation initiation factor 2-alpha (eIF2α). Phosphorylation of eIF2α is directly related to the dynamics of stress granules (SGs), which are membraneless organelles composed of RNA-binding proteins and mRNA. SGs play a critical role in mRNA metabolism and translational control. Other translation factors are also linked to cellular pathways, including SG dynamics following a stroke. Because the formation of SGs is closely connected to mRNA translation, it is interesting to study the relationship between SG dynamics and cellular outcome in cases of ischemic damage. Therefore, in this review, we focus on the role of SG dynamics during cerebral ischemia.
Collapse
|
29
|
Bal SS, Chen K, Yang FPG, Peng G. Arterial Input Function segmentation based on a contour geodesic model for tissue at risk identification in Ischemic Stroke. Med Phys 2022; 49:2475-2485. [DOI: 10.1002/mp.15508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 11/10/2022] Open
Affiliation(s)
- Sukhdeep Singh Bal
- Department of Mathematical sciences University of Liverpool Liverpool UK
- Center for Cognition and Mind Sciences National Tsing Hua University Taiwan
- International Intercollegiate Ph.D. Programme National Tsing Hua University Taiwan
| | - Ke Chen
- Department of Mathematical sciences University of Liverpool Liverpool Merseyside UK
| | - Fan Pei Gloria Yang
- Center for Cognition and Mind Sciences National Tsing Hua University Taiwan
- Department of Foreign Languages and Literature National Tsing Hua University Taiwan
- Department of Radiology Graduate School of Dentistry Osaka University Japan
- No. 101, Section 2, Guangfu Road, East District Hsinchu City 300 Taiwan
| | - Giia‐Sheun Peng
- Department of Neurology Tri‐Service General Hospital National Defense Medical Center Taipei Taiwan
- Division of Neurology Department of Internal Medicine Taipei Veterans General Hospital Hsinchu Branch Hsinchu County Taiwan
| |
Collapse
|
30
|
Post-stroke treatment with argon preserved neurons and attenuated microglia/macrophage activation long-termly in a rat model of transient middle cerebral artery occlusion (tMCAO). Sci Rep 2022; 12:691. [PMID: 35027642 PMCID: PMC8758662 DOI: 10.1038/s41598-021-04666-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 12/24/2021] [Indexed: 11/18/2022] Open
Abstract
In a previous study from our group, argon has shown to significantly attenuate brain injury, reduce brain inflammation and enhance M2 microglia/macrophage polarization until 7 days after ischemic stroke. However, the long-term effects of argon have not been reported thus far. In the present study, we analyzed the underlying neuroprotective effects and potential mechanisms of argon, up to 30 days after ischemic stroke. Argon administration with a 3 h delay after stroke onset and 1 h after reperfusion demonstrated long-term neuroprotective effect by preserving the neurons at the ischemic boundary zone 30 days after stroke. Furthermore, the excessive microglia/macrophage activation in rat brain was reduced by argon treatment 30 days after ischemic insult. However, long-lasting neurological improvement was not detectable. More sensorimotor functional measures, age- and disease-related models, as well as further histological and molecular biological analyses will be needed to extend the understanding of argon’s neuroprotective effects and mechanism of action after ischemic stroke.
Collapse
|
31
|
Desai SM, Jha RM, Linfante I. Collateral Circulation Augmentation and Neuroprotection as Adjuvant to Mechanical Thrombectomy in Acute Ischemic Stroke. Neurology 2021; 97:S178-S184. [PMID: 34785616 DOI: 10.1212/wnl.0000000000012809] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/03/2021] [Indexed: 01/22/2023] Open
Abstract
PURPOSE OF THE REVIEW Mechanical thrombectomy (MT)-mediated endovascular recanalization has dramatically transformed treatment and outcomes after acute ischemic stroke caused by a large vessel occlusion (LVO). Current guidelines recommend MT up to 24 hours from stroke onset in carefully selected patients based on favorable clinical and imaging parameters. Despite optimal patient selection and low complication rates with current recanalization technology, approximately 1 in 2 patients with LVO stroke do not achieve functional independence at 3 months. This ceiling effect of MT efficacy may be explained by ischemic core expansion into the ischemic penumbra before recanalization and neuronal loss occurring after recanalization. Factors affecting the efficacy of MT, or the degree of irreversible injury, include time from symptom onset to recanalization, collateral circulation status, and differences in neuronal vulnerability. The purpose of this brief review is to discuss potential targets for neuroprotection, present and future potential pharmacologic and nonpharmacologic agents, and the data available in the literature. RECENT FINDINGS In experimental ischemia models, several authors reported that pharmacologic and nonpharmacologic agents are able to slow the progression of ischemic core expansion. However, in the era of unsuccessful recanalization of the occluded artery, several neuroprotective agents that were promising in the preclinical stage failed phase II/III clinical trials. SUMMARY Providing neuroprotection before and after recanalization of an LVO may play an important role in improving outcomes in the era of MT. Neuroprotection is classically defined as a process that results in the salvage, recovery, or regeneration of neuronal (and other supporting CNS cell) structure or function. The advent of successful recanalization of acute LVO by MT in the majority of patients may spur the growth of effective neuroprotection.
Collapse
Affiliation(s)
- Shashvat M Desai
- From the Barrow Neurological Institute (S.M.D.), Department of Neurology, Phoenix, AZ; and Baptist Cardiac and Vascular Institute, Department of Neurology, Miami, FL
| | - Ruchira M Jha
- From the Barrow Neurological Institute (S.M.D.), Department of Neurology, Phoenix, AZ; and Baptist Cardiac and Vascular Institute, Department of Neurology, Miami, FL
| | - Italo Linfante
- From the Barrow Neurological Institute (S.M.D.), Department of Neurology, Phoenix, AZ; and Baptist Cardiac and Vascular Institute, Department of Neurology, Miami, FL.
| |
Collapse
|
32
|
Zhao HT, Tuohy MC, Chow D, Kozberg MG, Kim SH, Shaik MA, Hillman EMC. Neurovascular dynamics of repeated cortical spreading depolarizations after acute brain injury. Cell Rep 2021; 37:109794. [PMID: 34610299 PMCID: PMC8590206 DOI: 10.1016/j.celrep.2021.109794] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/30/2021] [Accepted: 09/14/2021] [Indexed: 11/30/2022] Open
Abstract
Cortical spreading depolarizations (CSDs) are increasingly suspected to play an exacerbating role in a range of acute brain injuries, including stroke, possibly through their interactions with cortical blood flow. We use simultaneous wide-field imaging of neural activity and hemodynamics in Thy1-GCaMP6f mice to explore the neurovascular dynamics of CSDs during and following Rose Bengal-mediated photothrombosis. CSDs are observed in all mice as slow-moving waves of GCaMP fluorescence extending far beyond the photothrombotic area. Initial CSDs are accompanied by profound vasoconstriction and leave residual oligemia and ischemia in their wake. Later, CSDs evoke variable responses, from constriction to biphasic to vasodilation. However, CSD-evoked vasoconstriction is found to be more likely during rapid, high-amplitude CSDs in regions with stronger oligemia and ischemia, which, in turn, worsens after each repeated CSD. This feedback loop may explain the variable but potentially devastating effects of CSDs in the context of acute brain injury. Zhao et al. use wide-field optical mapping of neuronal and hemodynamic activity in mice, capturing CSDs immediately following photothrombosis. Initial CSDs are accompanied by strong vasoconstriction, leaving persistent oligemia and ischemia. Region-dependent neurovascular responses to subsequent CSDs demonstrate a potential vicious cycle of CSD-dependent damage in acute brain injury.
Collapse
Affiliation(s)
- Hanzhi T Zhao
- Laboratory for Functional Optical Imaging, Mortimer B. Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Mary Claire Tuohy
- Laboratory for Functional Optical Imaging, Mortimer B. Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Daniel Chow
- Laboratory for Functional Optical Imaging, Mortimer B. Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Mariel G Kozberg
- Laboratory for Functional Optical Imaging, Mortimer B. Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Sharon H Kim
- Laboratory for Functional Optical Imaging, Mortimer B. Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Mohammed A Shaik
- Laboratory for Functional Optical Imaging, Mortimer B. Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA
| | - Elizabeth M C Hillman
- Laboratory for Functional Optical Imaging, Mortimer B. Zuckerman Mind Brain Behavior Institute, Departments of Biomedical Engineering and Radiology, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
33
|
Lu J, Mei Q, Hou X, Manaenko A, Zhou L, Liebeskind DS, Zhang JH, Li Y, Hu Q. Imaging Acute Stroke: From One-Size-Fit-All to Biomarkers. Front Neurol 2021; 12:697779. [PMID: 34630278 PMCID: PMC8497192 DOI: 10.3389/fneur.2021.697779] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/30/2021] [Indexed: 12/27/2022] Open
Abstract
In acute stroke management, time window has been rigidly used as a guide for decades and the reperfusion treatment is only available in the first few limited hours. Recently, imaging-based selection of patients has successfully expanded the treatment window out to 16 and even 24 h in the DEFUSE 3 and DAWN trials, respectively. Recent guidelines recommend the use of imaging techniques to guide therapeutic decision-making and expanded eligibility in acute ischemic stroke. A tissue window is proposed to replace the time window and serve as the surrogate marker for potentially salvageable tissue. This article reviews the evolution of time window, addresses the advantage of a tissue window in precision medicine for ischemic stroke, and discusses both the established and emerging techniques of neuroimaging and their roles in defining a tissue window. We also emphasize the metabolic imaging and molecular imaging of brain pathophysiology, and highlight its potential in patient selection and treatment response prediction in ischemic stroke.
Collapse
Affiliation(s)
- Jianfei Lu
- Central Laboratory, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiyong Mei
- Department of Neurosurgery, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Xianhua Hou
- Department of Neurology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Anatol Manaenko
- National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lili Zhou
- Department of Neurology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - David S. Liebeskind
- Neurovascular Imaging Research Core and University of California Los Angeles Stroke Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - John H. Zhang
- Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Yao Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Qin Hu
- Central Laboratory, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
Zhang H, Lin S, McElroy CL, Wang B, Jin D, Uteshev VV, Jin K. Circulating Pro-Inflammatory Exosomes Worsen Stroke Outcomes in Aging. Circ Res 2021; 129:e121-e140. [PMID: 34399581 DOI: 10.1161/circresaha.121.318897] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Hongxia Zhang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas
| | - Siyang Lin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas
| | - Christopher L McElroy
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas
| | - Brian Wang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas
| | - Dana Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas
| | - Victor V Uteshev
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
35
|
Muddasani V, de Havenon A, McNally JS, Baradaran H, Alexander MD. MR Perfusion in the Evaluation of Mechanical Thrombectomy Candidacy. Top Magn Reson Imaging 2021; 30:197-204. [PMID: 34397969 PMCID: PMC8371677 DOI: 10.1097/rmr.0000000000000277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
ABSTRACT Stroke is a leading cause of disability and mortality, and the incidence of ischemic stroke is projected to continue to rise in coming decades. These projections emphasize the need for improved imaging techniques for accurate diagnosis allowing effective treatments for ischemic stroke. Ischemic stroke is commonly evaluated with computed tomography (CT) or magnetic resonance imaging (MRI). Noncontrast CT is typically used within 4.5 hours of symptom onset to identify candidates for thrombolysis. Beyond this time window, thrombolytic therapy may lead to poor outcomes if patients are not optimally selected using appropriate imaging. MRI provides an accurate method for the earliest identification of core infarct, and MR perfusion can identify salvageable hypoperfused penumbra. The prognostic value for a better outcome in these patients lies in the ability to distinguish between core infarct and salvageable brain at risk-the ischemic penumbra-which is a function of the degree of ischemia and time. Many centers underutilize MRI for acute evaluation of ischemic stroke. This review will illustrate how perfusion-diffusion mismatch calculated from diffusion-weighted MRI and MR perfusion is a reliable approach for patient selection for stroke therapy and can be performed in timeframes that are comparable to CT-based algorithms while providing potentially superior diagnostic information.
Collapse
Affiliation(s)
| | - Adam de Havenon
- Department of Neurology, University of Utah, Salt Lake City, UT
| | - J Scott McNally
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT
| | - Hediyeh Baradaran
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT
| | - Matthew D Alexander
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT
- Department of Neurosurgery, University of Utah, Salt Lake City, UT
| |
Collapse
|
36
|
Four Decades of Ischemic Penumbra and Its Implication for Ischemic Stroke. Transl Stroke Res 2021; 12:937-945. [PMID: 34224106 DOI: 10.1007/s12975-021-00916-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 12/15/2022]
Abstract
The ischemic penumbra defined four decades ago has been the main battleground of ischemic stroke. The evolving ischemic penumbra concept has been providing insight for the development of vascular and cellular approaches as well as diagnostic tools for the treatment of ischemic stroke. rt-PA thrombolytic therapy to prevent the transition of ischemic penumbra to core has been approved for acute ischemic stroke within 3 h and was later recommended to extend to 4.5 h after symptom onset. Mechanical thrombectomy was introduced for the treatment of acute ischemic stroke with a therapeutic window of up to 24 h after stroke onset. Multiple modalities brain imaging techniques have been developed that provide guidance to define ischemic penumbra for reperfusion therapy in clinical practice. Cellular and molecular dissection of ischemic penumbra has been providing targets for the development of neuroprotective therapy for ischemic stroke. However, the dynamic nature of ischemic penumbra implicates that infarct core eventually expands into penumbra over time without reperfusion, dictating relative short therapeutic windows and limiting the impact of current reperfusion intervention. Entering the 5th decade since the introduction, ischemic penumbra remains the main focus of ischemic stroke research and clinical practice. In this review, we summarized the evolving ischemic penumbra concept and its implication in the development of vascular and cellular interventions as well as diagnostic tools for acute ischemic stroke. In addition, we discussed future perspectives on expansion of the campaign beyond ischemic penumbra to develop treatment for ischemic stroke.
Collapse
|
37
|
Assessing the Potential of Molecular Imaging for Myelin Quantification in Organotypic Cultures. Pharmaceutics 2021; 13:pharmaceutics13070975. [PMID: 34203246 PMCID: PMC8309097 DOI: 10.3390/pharmaceutics13070975] [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: 05/14/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 11/17/2022] Open
Abstract
Ex vivo models for the noninvasive study of myelin-related diseases represent an essential tool to understand the mechanisms of diseases and develop therapies against them. Herein, we assessed the potential of multimodal imaging traceable myelin-targeting liposomes to quantify myelin in organotypic cultures. Methods: MRI testing was used to image mouse cerebellar tissue sections and organotypic cultures. Demyelination was induced by lysolecithin treatment. Myelin-targeting liposomes were synthetized and characterized, and their capacity to quantify myelin was tested by fluorescence imaging. Results: Imaging of freshly excised tissue sections ranging from 300 µm to 1 mm in thickness was achieved with good contrast between white (WM) and gray matter (GM) using T2w MRI. The typical loss of stiffness, WM structures, and thickness of organotypic cultures required the use of diffusion-weighted methods. Designed myelin-targeting liposomes allowed for semiquantitative detection by fluorescence, but the specificity for myelin was not consistent between assays due to the unspecific binding of liposomes. Conclusions: With respect to the sensitivity, imaging of brain tissue sections and organotypic cultures by MRI is feasible, and myelin-targeting nanosystems are a promising solution to quantify myelin ex vivo. With respect to specificity, fine tuning of the probe is required. Lipid-based systems may not be suitable for this goal, due to unspecific binding to tissues.
Collapse
|
38
|
Iglesias-Rey R, da Silva-Candal A, Rodríguez-Yáñez M, Estany-Gestal A, Regueiro U, Maqueda E, Ávila-Gómez P, Pumar JM, Castillo J, Sobrino T, Campos F, Hervella P. Neurological Instability in Ischemic Stroke: Relation with Outcome, Latency Time, and Molecular Markers. Transl Stroke Res 2021; 13:228-237. [PMID: 34165728 PMCID: PMC8918467 DOI: 10.1007/s12975-021-00924-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 01/08/2023]
Abstract
The National Institutes of Health Stroke Scale (NIHSS) is commonly used to evaluate stroke neurological deficits and to predict the patient’s outcome. Neurological instability (NI), defined as the variation of the NIHSS in the first 48 h, is a simple clinical metric that reflects dynamic changes in the area of the brain affected by the ischemia. We hypothesize that NI may represent areas of cerebral instability known as penumbra, which could expand or reduce brain injury and its associated neurological sequels. In this work, our aim was to analyze the association of NI with the functional outcome at 3 months and to study clinical biomarkers associated to NI as surrogate biomarkers of ischemic and inflammatory penumbrae in ischemic stroke (IS) patients. We included 663 IS patients in a retrospective observational study. Neutral NI was defined as a variation in the NI scale between − 5 and 5% (37.1%). Positive NI is attributed to patients with an improvement of > 5% NI after 48 h (48.9%), while negative NI is assigned to patients values lower than − 5% (14.0%). Poor outcome was assigned to patients with mRS ≥ 3 at 3 months. We observed an inverse association of poor outcome with positive NI (OR, 0.35; 95%CI, 0.18–0.67; p = 0.002) and a direct association with negative NI (OR, 6.30; 95%CI, 2.12–18.65; p = 0.001). Negative NI showed a higher association with poor outcome than most clinical markers. Regarding good functional outcome, positive NI was the marker with the higher association (19.31; CI 95%, 9.03–41.28; p < 0.0001) and with the highest percentage of identified patients with good functional outcome (17.6%). Patients with negative NI have higher glutamate levels compared with patients with neutral and positive NI (p < 0.0001). IL6 levels are significantly lower in patients with positive NI compared with neutral NI (p < 0.0001), while patients with negative NI showed the highest IL6 values (p < 0.0001). High glutamate levels were associated with negative NI at short latency times, decreasing at higher latency times. An opposite trend was observed for inflammation, and IL6 levels were similar in patients with positive and negative NI in the first 6 h and then higher in patients with negative NI. These results support NI as a prognosis factor in IS and the hypothesis of the existence of a delayed inflammatory penumbra, opening up the possibility of extending the therapeutic window for IS.
Collapse
Affiliation(s)
- Ramón Iglesias-Rey
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain.
| | - Andres da Silva-Candal
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Manuel Rodríguez-Yáñez
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, Santiago de Compostela, Spain
| | - Ana Estany-Gestal
- Unit of Methodology of the Research, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Uxía Regueiro
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Elena Maqueda
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Paulo Ávila-Gómez
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - José Manuel Pumar
- Department of Neuroradiology, Hospital Clínico Universitario, Santiago de Compostela, Spain
| | - José Castillo
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Pablo Hervella
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain.
| |
Collapse
|
39
|
Michinaga S, Koyama Y. Pathophysiological Responses and Roles of Astrocytes in Traumatic Brain Injury. Int J Mol Sci 2021; 22:ijms22126418. [PMID: 34203960 PMCID: PMC8232783 DOI: 10.3390/ijms22126418] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is immediate damage caused by a blow to the head resulting from traffic accidents, falls, and sporting activity, which causes death or serious disabilities in survivors. TBI induces multiple secondary injuries, including neuroinflammation, disruption of the blood–brain barrier (BBB), and brain edema. Despite these emergent conditions, current therapies for TBI are limited or insufficient in some cases. Although several candidate drugs exerted beneficial effects in TBI animal models, most of them failed to show significant effects in clinical trials. Multiple studies have suggested that astrocytes play a key role in the pathogenesis of TBI. Increased reactive astrocytes and astrocyte-derived factors are commonly observed in both TBI patients and experimental animal models. Astrocytes have beneficial and detrimental effects on TBI, including promotion and restriction of neurogenesis and synaptogenesis, acceleration and suppression of neuroinflammation, and disruption and repair of the BBB via multiple bioactive factors. Additionally, astrocytic aquaporin-4 is involved in the formation of cytotoxic edema. Thus, astrocytes are attractive targets for novel therapeutic drugs for TBI, although astrocyte-targeting drugs have not yet been developed. This article reviews recent observations of the roles of astrocytes and expected astrocyte-targeting drugs in TBI.
Collapse
Affiliation(s)
- Shotaro Michinaga
- Department of Pharmacodynamics, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan;
| | - Yutaka Koyama
- Laboratory of Pharmacology, Kobe Pharmaceutical University, 4-19-1 Motoyama-Kita Higashinada, Kobe 668-8558, Japan
- Correspondence: ; Tel.: +81-78-441-7572
| |
Collapse
|
40
|
Bai R, Lang Y, Shao J, Deng Y, Refuhati R, Cui L. The Role of NLRP3 Inflammasome in Cerebrovascular Diseases Pathology and Possible Therapeutic Targets. ASN Neuro 2021; 13:17590914211018100. [PMID: 34053242 PMCID: PMC8168029 DOI: 10.1177/17590914211018100] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cerebrovascular diseases are pathological conditions involving impaired blood flow in the brain, primarily including ischaemic stroke, intracranial haemorrhage, and subarachnoid haemorrhage. The nucleotide-binding and oligomerisation (NOD) domain-like receptor (NLR) family pyrin domain (PYD)-containing 3 (NLRP3) inflammasome is a protein complex and a vital component of the immune system. Emerging evidence has indicated that the NLRP3 inflammasome plays an important role in cerebrovascular diseases. The function of the NLRP3 inflammasome in the pathogenesis of cerebrovascular diseases remains an interesting field of research. In this review, we first summarised the pathological mechanism of cerebrovascular diseases and the pathological mechanism of the NLRP3 inflammasome in aggravating atherosclerosis and cerebrovascular diseases. Second, we outlined signalling pathways through which the NLRP3 inflammasome participates in aggravating or mitigating cerebrovascular diseases. Reactive oxygen species (ROS)/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), ROS/thioredoxin-interacting protein (TXNIP) and purinergic receptor-7 (P2X7R) signalling pathways can activate the NLRP3 inflammasome; activation of the NLRP3 inflammasome can aggravate cerebrovascular diseases by mediating apoptosis and pyroptosis. Autophagy/mitochondrial autophagy, nuclear factor E2-related factor-2 (Nrf2), interferon (IFN)-β, sirtuin (SIRT), and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) reportedly alleviate cerebrovascular diseases by inhibiting NLRP3 inflammasome activation. Finally, we explored specific inhibitors of the NLRP3 inflammasome based on the two-step activation of the NLRP3 inflammasome, which can be developed as new drugs to treat cerebrovascular diseases.
Collapse
Affiliation(s)
- Rongrong Bai
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yue Lang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jie Shao
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yu Deng
- Department of Hepatopancreatobiliary Surgery, The First Hospital of Jilin University, Changchun, China
| | - Reyisha Refuhati
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Li Cui
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
41
|
Dysfunction of the Neurovascular Unit in Ischemic Stroke: Highlights on microRNAs and Exosomes as Potential Biomarkers and Therapy. Int J Mol Sci 2021; 22:ijms22115621. [PMID: 34070696 PMCID: PMC8198979 DOI: 10.3390/ijms22115621] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/21/2021] [Accepted: 05/23/2021] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke is a damaging cerebral vascular disease associated with high disability and mortality rates worldwide. In spite of the continuous development of new diagnostic and prognostic methods, early detection and outcome prediction are often very difficult. The neurovascular unit (NVU) is a complex multicellular entity linking the interactions between neurons, glial cells, and brain vessels. Novel research has revealed that exosome-mediated transfer of microRNAs plays an important role in cell-to-cell communication and, thus, is integral in the multicellular crosstalk within the NVU. After a stroke, NVU homeostasis is altered, which induces the release of several potential biomarkers into the blood vessels. The addition of biological data representing all constituents of the NVU to clinical and neuroradiological findings can significantly advance stroke evaluation and prognosis. In this review, we present the current literature regarding the possible beneficial roles of exosomes derived from the components of the NVU and multipotent mesenchymal stem cells in preclinical studies of ischemic stroke. We also discuss the most relevant clinical trials on the diagnostic and prognostic roles of exosomes in stroke patients.
Collapse
|
42
|
New Approaches in Nanomedicine for Ischemic Stroke. Pharmaceutics 2021; 13:pharmaceutics13050757. [PMID: 34065179 PMCID: PMC8161190 DOI: 10.3390/pharmaceutics13050757] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 12/20/2022] Open
Abstract
Ischemic stroke, caused by the interruption of blood flow to the brain and subsequent neuronal death, represents one of the main causes of disability in developed countries. Therapeutic methods such as recanalization approaches, neuroprotective drugs, or recovery strategies have been widely developed to improve the patient's outcome; however, important limitations such as a narrow therapeutic window, the ability to reach brain targets, or drug side effects constitute some of the main aspects that limit the clinical applicability of the current treatments. Nanotechnology has emerged as a promising tool to overcome many of these drug limitations and improve the efficacy of treatments for neurological diseases such as stroke. The use of nanoparticles as a contrast agent or as drug carriers to a specific target are some of the most common approaches developed in nanomedicine for stroke. Throughout this review, we have summarized our experience of using nanotechnology tools for the study of stroke and the search for novel therapies.
Collapse
|
43
|
Patabendige A, Singh A, Jenkins S, Sen J, Chen R. Astrocyte Activation in Neurovascular Damage and Repair Following Ischaemic Stroke. Int J Mol Sci 2021; 22:4280. [PMID: 33924191 PMCID: PMC8074612 DOI: 10.3390/ijms22084280] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/11/2021] [Accepted: 04/15/2021] [Indexed: 12/11/2022] Open
Abstract
Transient or permanent loss of tissue perfusion due to ischaemic stroke can lead to damage to the neurovasculature, and disrupt brain homeostasis, causing long-term motor and cognitive deficits. Despite promising pre-clinical studies, clinically approved neuroprotective therapies are lacking. Most studies have focused on neurons while ignoring the important roles of other cells of the neurovascular unit, such as astrocytes and pericytes. Astrocytes are important for the development and maintenance of the blood-brain barrier, brain homeostasis, structural support, control of cerebral blood flow and secretion of neuroprotective factors. Emerging data suggest that astrocyte activation exerts both beneficial and detrimental effects following ischaemic stroke. Activated astrocytes provide neuroprotection and contribute to neurorestoration, but also secrete inflammatory modulators, leading to aggravation of the ischaemic lesion. Astrocytes are more resistant than other cell types to stroke pathology, and exert a regulative effect in response to ischaemia. These roles of astrocytes following ischaemic stroke remain incompletely understood, though they represent an appealing target for neurovascular protection following stroke. In this review, we summarise the astrocytic contributions to neurovascular damage and repair following ischaemic stroke, and explore mechanisms of neuroprotection that promote revascularisation and neurorestoration, which may be targeted for developing novel therapies for ischaemic stroke.
Collapse
Affiliation(s)
- Adjanie Patabendige
- Brain Barriers Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2321, Australia;
- Priority Research Centre for Stroke and Brain Injury, and Priority Research Centre for Brain & Mental Health, University of Newcastle, Callaghan, NSW 2321, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
- Institute of Infection & Global Health, University of Liverpool, Liverpool L7 3EA, UK
| | - Ayesha Singh
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK;
| | - Stuart Jenkins
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK; (S.J.); (J.S.)
- Neural Tissue Engineering: Keele (NTEK), Keele University, Staffordshire ST5 5BG, UK
| | - Jon Sen
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK; (S.J.); (J.S.)
- Clinical Informatics and Neurosurgery Fellow, The Cleveland Clinic, 33 Grosvenor Square, London SW1X 7HY, UK
| | - Ruoli Chen
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK;
| |
Collapse
|
44
|
Krag AE, Blauenfeldt RA. Fibrinolysis and Remote Ischemic Conditioning: Mechanisms and Treatment Perspectives in Stroke. Semin Thromb Hemost 2021; 47:610-620. [PMID: 33878783 DOI: 10.1055/s-0041-1725095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Stroke is a leading cause of death and disability. Intravenous thrombolysis and mechanical thrombectomy have greatly improved outcomes in acute ischemic stroke (AIS). However, only a minority of patients receive reperfusion therapies, highlighting the need for novel neuroprotective therapies. Remote ischemic conditioning (RIC), consisting of brief, intermittent extremity occlusion and reperfusion induced with an inflatable cuff, is a potential neuroprotective therapy in acute stroke. The objective of this narrative review is to describe the effect of RIC on endogenous fibrinolysis and, from this perspective, investigate the potential of RIC in the prevention and treatment of stroke. A systematic literature search was performed in PubMed, and human studies in English were included. Seven studies had investigated the effect of RIC on fibrinolysis in humans. Long-term daily administration of RIC increased endogenous fibrinolysis, whereas a single RIC treatment did not acutely influence endogenous fibrinolysis. Fifteen studies had investigated the effect of RIC as a neuroprotective therapy in the prevention and treatment of stroke. Long-term RIC administration proved effective in reducing new cerebral vascular lesions in patients with established cerebrovascular disease. In patients with acute stroke, RIC was safe and feasible, though its clinical efficacy as a neuroprotectant is yet unproven. In conclusion, a single RIC treatment does not affect fibrinolysis in the acute phase, whereas long-term RIC administration may increase endogenous fibrinolysis. Increased endogenous fibrinolysis is unlikely to be the mediator of the acute neuroprotective effect of RIC in stroke patients, whereas it may partly explain the reduced stroke recurrence associated with long-term RIC treatment.
Collapse
Affiliation(s)
- Andreas Engel Krag
- Thrombosis and Hemostasis Research Unit, Aarhus University Hospital, Aarhus, Denmark
| | - Rolf Ankerlund Blauenfeldt
- Department of Neurology, Danish Stroke Center, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
45
|
Pires Monteiro S, Voogd E, Muzzi L, De Vecchis G, Mossink B, Levers M, Hassink G, Van Putten M, Le Feber J, Hofmeijer J, Frega M. Neuroprotective effect of hypoxic preconditioning and neuronal activation in a in vitro human model of the ischemic penumbra. J Neural Eng 2021; 18:036016. [PMID: 33724235 DOI: 10.1088/1741-2552/abe68a] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE In ischemic stroke, treatments to protect neurons from irreversible damage are urgently needed. Studies in animal models have shown that neuroprotective treatments targeting neuronal silencing improve brain recovery, but in clinical trials none of these were effective in patients. This failure of translation poses doubts on the real efficacy of treatments tested and on the validity of animal models for human stroke. Here, we established a human neuronal model of the ischemic penumbra by using human induced pluripotent stem cells and we provided an in-depth characterization of neuronal responses to hypoxia and treatment strategies at the network level. APPROACH We generated neurons from induced pluripotent stem cells derived from healthy donor and we cultured them on micro-electrode arrays. We measured the electrophysiological activity of human neuronal networks under controlled hypoxic conditions. We tested the effect of different treatment strategies on neuronal network functionality. MAIN RESULTS Human neuronal networks are vulnerable to hypoxia reflected by a decrease in activity and synchronicity under low oxygen conditions. We observe that full, partial or absent recovery depend on the timing of re-oxygenation and we provide a critical time threshold that, if crossed, is associated with irreversible impairments. We found that hypoxic preconditioning improves resistance to a second hypoxic insult. Finally, in contrast to previously tested, ineffective treatments, we show that stimulatory treatments counteracting neuronal silencing during hypoxia, such as optogenetic stimulation, are neuroprotective. SIGNIFICANCE We presented a human neuronal model of the ischemic penumbra and we provided insights that may offer the basis for novel therapeutic approaches for patients after stroke. The use of human neurons might improve drug discovery and translation of findings to patients and might open new perspectives for personalized investigations.
Collapse
Affiliation(s)
- Sara Pires Monteiro
- Department of Clinical Neurophysiology, University of Twente, 7522 NB Enschede, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Thomson J, New PW. Pharmacological management of stroke in older people. JOURNAL OF PHARMACY PRACTICE AND RESEARCH 2021. [DOI: 10.1002/jppr.1703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Julia Thomson
- Department of Medicine Rehabilitation and Aged Services Program, Kingston Centre Monash Health Clayton Australia
| | - Peter Wayne New
- Department of Medicine Rehabilitation and Aged Services Program, Kingston Centre Monash Health Clayton Australia
- Rehabilitation Services Caulfield Hospital Alfred Health Victoria Australia
- Epworth‐Monash Rehabilitation Medicine Unit Monash Medical School Monash University Parkville Australia
- Department of Epidemiology and Preventive Medicine School of Public Health and Preventive Medicine Monash University Parkville Australia
| |
Collapse
|
47
|
Tchantchou F, Miller C, Goodfellow M, Puche A, Fiskum G. Hypobaria-Induced Oxidative Stress Facilitates Homocysteine Transsulfuration and Promotes Glutathione Oxidation in Rats with Mild Traumatic Brain Injury. J Cent Nerv Syst Dis 2021; 13:1179573520988193. [PMID: 33597815 PMCID: PMC7863175 DOI: 10.1177/1179573520988193] [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] [Received: 07/28/2020] [Accepted: 12/18/2020] [Indexed: 01/14/2023] Open
Abstract
Background: United States service members injured in combat theatre are often aeromedically evacuated within a few days to regional military hospitals. Animal and epidemiological research indicates that early exposure to flight hypobaria may worsen brain and other injuries. The mechanisms by which secondary exposure to hypobaria worsen trauma outcomes are not well elucidated. This study tested the hypothesis that hypobaria-induced oxidative stress and associated changes in homocysteine levels play a role in traumatic brain injury (TBI) pathological progression caused by hypobaria. Methods: Male Sprague Dawley rats were exposed to a 6 h hypobaria 24 h after mild TBI by the controlled cortical impact. Plasma and brain tissues were assessed for homocysteine levels, oxidative stress markers or glutathione metabolism, and behavioral deficits post-injury in the absence and presence of hypobaria exposure. Results: We found that hypobaria after TBI increased oxidative stress markers, altered homocysteine metabolism, and promoted glutathione oxidation. Increased glutathione metabolism was driven by differential upregulation of glutathione metabolizing genes. These changes correlated with increased anxiety-like behavior. Conclusion: These data provide evidence that hypobaria exposure after TBI increases oxidative stress and alters homocysteine elimination likely through enhanced glutathione metabolism. This pathway may represent a compensatory mechanism to attenuate free radical formation. Thus, hypobaria-induced enhancement of glutathione metabolism represents a potential therapeutic target for TBI management.
Collapse
Affiliation(s)
- Flaubert Tchantchou
- Department of Anesthesiology and the Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, USA
| | - Catriona Miller
- Aeromedical Research, U.S Air Force School of Aerospace Medicine, Wright-Patterson, OH, USA
| | - Molly Goodfellow
- Department of Anesthesiology and the Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, USA
| | - Adam Puche
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, USA
| | - Gary Fiskum
- Department of Anesthesiology and the Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, USA
| |
Collapse
|
48
|
Lin L, Yang J, Chen C, Tian H, Bivard A, Spratt NJ, Levi CR, Parsons MW. Association of Collateral Status and Ischemic Core Growth in Patients With Acute Ischemic Stroke. Neurology 2020; 96:e161-e170. [PMID: 33262233 DOI: 10.1212/wnl.0000000000011258] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 08/12/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To test the hypothesis that patients with acute ischemic stroke with poorer collaterals would have faster ischemic core growth, we included 2 cohorts in the study: cohort 1 of 342 patients for derivation and cohort 2 of 414 patients for validation. METHODS Patients with acute ischemic stroke with large vessel occlusion were included. Core growth rate was calculated by the following equation: core growth rate = acute core volume on CT perfusion (CTP)/time from stroke onset to CTP. Collateral status was assessed by the ratio of severe hypoperfusion volume within the hypoperfusion region of CTP. The CTP collateral index was categorized in tertiles; for each tertile, core growth rate was summarized as median and interquartile range. Simple linear regressions were then performed to measure the predictive power of CTP collateral index in core growth rate. RESULTS For patients allocated to good collateral on CTP (tertile 1 of collateral index), moderate collateral (tertile 2), and poor collateral (tertile 3), the median core growth rate was 2.93 mL/h (1.10-7.94), 8.65 mL/h (4.53-18.13), and 25.41 mL/h (12.83-45.07), respectively. Increments in the collateral index by 1% resulted in an increase of core growth by 0.57 mL/h (coefficient 0.57, 95% confidence interval [0.46, 0.68], p < 0.001). The relationship of core growth and CTP collateral index was validated in cohort 2. An increment in collateral index by 1% resulted in an increase of core growth by 0.59 mL/h (coefficient 0.59 [0.48-0.71], p < 0.001) in cohort 2. CONCLUSION Collateral status is a major determinant of ischemic core growth.
Collapse
Affiliation(s)
- Longting Lin
- From the Department of Neurology (L.L., J.Y.), Ningbo First Hospital, China; School of Medicine and Public Health (L.L., C.C., H.T., N.J.S., C.R.L., M.W.P.), University of Newcastle; Melbourne Brain Centre at Royal Melbourne Hospital (A.B., M.W.P.), University of Melbourne; and The Sydney Partnership for Health, Education, Research and Enterprise (C.R.L.), Australia
| | - Jianhong Yang
- From the Department of Neurology (L.L., J.Y.), Ningbo First Hospital, China; School of Medicine and Public Health (L.L., C.C., H.T., N.J.S., C.R.L., M.W.P.), University of Newcastle; Melbourne Brain Centre at Royal Melbourne Hospital (A.B., M.W.P.), University of Melbourne; and The Sydney Partnership for Health, Education, Research and Enterprise (C.R.L.), Australia
| | - Chushuang Chen
- From the Department of Neurology (L.L., J.Y.), Ningbo First Hospital, China; School of Medicine and Public Health (L.L., C.C., H.T., N.J.S., C.R.L., M.W.P.), University of Newcastle; Melbourne Brain Centre at Royal Melbourne Hospital (A.B., M.W.P.), University of Melbourne; and The Sydney Partnership for Health, Education, Research and Enterprise (C.R.L.), Australia
| | - Huiqiao Tian
- From the Department of Neurology (L.L., J.Y.), Ningbo First Hospital, China; School of Medicine and Public Health (L.L., C.C., H.T., N.J.S., C.R.L., M.W.P.), University of Newcastle; Melbourne Brain Centre at Royal Melbourne Hospital (A.B., M.W.P.), University of Melbourne; and The Sydney Partnership for Health, Education, Research and Enterprise (C.R.L.), Australia
| | - Andrew Bivard
- From the Department of Neurology (L.L., J.Y.), Ningbo First Hospital, China; School of Medicine and Public Health (L.L., C.C., H.T., N.J.S., C.R.L., M.W.P.), University of Newcastle; Melbourne Brain Centre at Royal Melbourne Hospital (A.B., M.W.P.), University of Melbourne; and The Sydney Partnership for Health, Education, Research and Enterprise (C.R.L.), Australia
| | - Neil J Spratt
- From the Department of Neurology (L.L., J.Y.), Ningbo First Hospital, China; School of Medicine and Public Health (L.L., C.C., H.T., N.J.S., C.R.L., M.W.P.), University of Newcastle; Melbourne Brain Centre at Royal Melbourne Hospital (A.B., M.W.P.), University of Melbourne; and The Sydney Partnership for Health, Education, Research and Enterprise (C.R.L.), Australia
| | - Christopher R Levi
- From the Department of Neurology (L.L., J.Y.), Ningbo First Hospital, China; School of Medicine and Public Health (L.L., C.C., H.T., N.J.S., C.R.L., M.W.P.), University of Newcastle; Melbourne Brain Centre at Royal Melbourne Hospital (A.B., M.W.P.), University of Melbourne; and The Sydney Partnership for Health, Education, Research and Enterprise (C.R.L.), Australia
| | - Mark W Parsons
- From the Department of Neurology (L.L., J.Y.), Ningbo First Hospital, China; School of Medicine and Public Health (L.L., C.C., H.T., N.J.S., C.R.L., M.W.P.), University of Newcastle; Melbourne Brain Centre at Royal Melbourne Hospital (A.B., M.W.P.), University of Melbourne; and The Sydney Partnership for Health, Education, Research and Enterprise (C.R.L.), Australia.
| | | |
Collapse
|
49
|
Paul S, Candelario-Jalil E. Emerging neuroprotective strategies for the treatment of ischemic stroke: An overview of clinical and preclinical studies. Exp Neurol 2020; 335:113518. [PMID: 33144066 DOI: 10.1016/j.expneurol.2020.113518] [Citation(s) in RCA: 404] [Impact Index Per Article: 80.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/20/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022]
Abstract
Stroke is the leading cause of disability and thesecond leading cause of death worldwide. With the global population aged 65 and over growing faster than all other age groups, the incidence of stroke is also increasing. In addition, there is a shift in the overall stroke burden towards younger age groups, particularly in low and middle-income countries. Stroke in most cases is caused due to an abrupt blockage of an artery (ischemic stroke), but in some instances stroke may be caused due to bleeding into brain tissue when a blood vessel ruptures (hemorrhagic stroke). Although treatment options for stroke are still limited, with the advancement in recanalization therapy using both pharmacological and mechanical thrombolysis some progress has been made in helping patients recover from ischemic stroke. However, there is still a substantial need for the development of therapeutic agents for neuroprotection in acute ischemic stroke to protect the brain from damage prior to and during recanalization, extend the therapeutic time window for intervention and further improve functional outcome. The current review has assessed the past challenges in developing neuroprotective strategies, evaluated the recent advances in clinical trials, discussed the recent initiative by the National Institute of Neurological Disorders and Stroke in USA for the search of novel neuroprotectants (Stroke Preclinical Assessment Network, SPAN) and identified emerging neuroprotectants being currently evaluated in preclinical studies. The underlying molecular mechanism of each of the neuroprotective strategies have also been summarized, which could assist in the development of future strategies for combinational therapy in stroke treatment.
Collapse
Affiliation(s)
- Surojit Paul
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
50
|
North K, Slayden A, Mysiewicz S, Bukiya A, Dopico A. Celastrol Dilates and Counteracts Ethanol-Induced Constriction of Cerebral Arteries. J Pharmacol Exp Ther 2020; 375:247-257. [PMID: 32862144 PMCID: PMC7589950 DOI: 10.1124/jpet.120.000152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 08/11/2020] [Indexed: 12/30/2022] Open
Abstract
The increasing recognition of the role played by cerebral artery dysfunction in brain disorders has fueled the search for new cerebrovascular dilators. Celastrol, a natural triterpene undergoing clinical trials for treating obesity, exerts neuroprotection, which was linked to its antioxidant/anti-inflammatory activities. We previously showed that celastrol fit pharmacophore criteria for activating calcium- and voltage-gated potassium channels of large conductance (BK channels) made of subunits cloned from cerebrovascular smooth muscle (SM). These recombinant BK channels expressed in a heterologous system were activated by celastrol. Activation of native SM BK channels is well known to evoke cerebral artery dilation. Current data demonstrate that celastrol (1-100 µM) dilates de-endothelialized, ex vivo pressurized middle cerebral arteries (MCAs) from rats, with EC50 = 45 µM and maximal effective concentration (Emax)= 100 µM and with MCA diameter reaching a 10% increase over vehicle-containing, time-matched values (P < 0.05). A similar vasodilatory efficacy is achieved when celastrol is probed on MCA segments with intact endothelium. Selective BK blocking with 1 μM paxilline blunts celastrol vasodilation. Similar blunting is achieved with 0.8 mM 4-aminopirydine, which blocks voltage-gated K+ channels other than BK. Using an in vivo rat cranial window, we further demonstrate that intracarotid injections of 45 μM celastrol into pial arteries branching from MCA mimics celastrol ex vivo action. MCA constriction by ethanol concentrations reached in blood during moderate-heavy alcohol drinking (50 mM), which involves SM BK inhibition, is both prevented and reverted by celastrol. We conclude that celastrol could be an effective cerebrovascular dilator and antagonist of alcohol-induced cerebrovascular constriction, with its efficacy being uncompromised by conditions that disrupt endothelial and/or BK function. SIGNIFICANCE STATEMENT: Our study demonstrates for the first time that celastrol significantly dilates rat cerebral arteries both ex vivo and in vivo and both prevents and reverses ethanol-induced cerebral artery constriction. Celastrol actions are endothelium-independent but mediated through voltage-gated (KV) and calcium- and voltage-gated potassium channel of large conductance (BK) K+ channels. This makes celastrol an appealing new agent to evoke cerebrovascular dilation under conditions in which endothelial and/or BK channel function are impaired.
Collapse
Affiliation(s)
- Kelsey North
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Alexandria Slayden
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Steven Mysiewicz
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Anna Bukiya
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Alex Dopico
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|