51
|
Goodman GW, Do TH, Tan C, Ritzel RM. Drivers of Chronic Pathology Following Ischemic Stroke: A Descriptive Review. Cell Mol Neurobiol 2023; 44:7. [PMID: 38112809 DOI: 10.1007/s10571-023-01437-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/25/2023] [Indexed: 12/21/2023]
Abstract
Stroke is the third leading cause of death and long-term disability in the world. Considered largely a disease of aging, its global economic and healthcare burden is expected to rise as more people survive into advanced age. With recent advances in acute stroke management, including the expansion of time windows for treatment with intravenous thrombolysis and mechanical thrombectomy, we are likely to see an increase in survival rates. It is therefore critically important to understand the complete pathophysiology of ischemic stroke, both in the acute and subacute stages and during the chronic phase in the months and years following an ischemic event. One of the most clinically relevant aspects of the chronic sequelae of stroke is its extended negative effect on cognition. Cognitive impairment may be related to the deterioration and dysfunctional reorganization of white matter seen at later timepoints after stroke, as well as ongoing progressive neurodegeneration. The vasculature of the brain also undergoes significant insult and remodeling following stroke, undergoing changes which may further contribute to chronic stroke pathology. While inflammation and the immune response are well established drivers of acute stroke pathology, the chronicity and functional role of innate and adaptive immune responses in the post-ischemic brain and in the peripheral environment remain largely uncharacterized. In this review, we summarize the current literature on post-stroke injury progression, its chronic pathological features, and the putative secondary injury mechanisms underlying the development of cognitive impairment and dementia. We present findings from clinical and experimental studies and discuss the long-term effects of ischemic stroke on both brain anatomy and functional outcome. Identifying mechanisms that occur months to years after injury could lead to treatment strategies in the chronic phase of stroke to help mitigate stroke-associated cognitive decline in patients.
Collapse
Affiliation(s)
- Grant W Goodman
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Trang H Do
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chunfeng Tan
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Rodney M Ritzel
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
52
|
Zhou J, Khateeb K, Yazdan-Shahmorad A. Early Intervention with Electrical Stimulation Reduces Neural Damage After Stroke in Non-human Primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.18.572235. [PMID: 38187669 PMCID: PMC10769281 DOI: 10.1101/2023.12.18.572235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Ischemic stroke is a neurological condition that results in significant mortality and long-term disability for adults, creating huge health burdens worldwide. For stroke patients, acute intervention offers the most critical therapeutic opportunity as it can reduce irreversible tissue injury and improve functional outcomes. However, currently available treatments within the acute window are highly limited. Although emerging neuromodulation therapies have been tested for chronic stroke patients, acute stimulation is rarely studied due to the risk of causing adverse effects related to ischemia-induced electrical instability. To address this gap, we combined electrophysiology and histology tools to investigate the effects of acute electrical stimulation on ischemic neural damage in non-human primates. Specifically, we induced photothrombotic lesions in the monkey sensorimotor cortex while collecting electrocorticography (ECoG) signals through a customized neural interface. Gamma activity in ECoG was used as an electrophysiological marker to track the effects of stimulation on neural activation. Meanwhile, histological analysis including Nissl, cFos, and microglial staining was performed to evaluate the tissue response to ischemic injury. Comparing stimulated monkeys to controls, we found that theta-burst stimulation administered directly adjacent to the ischemic infarct at 1 hour post-stroke briefly inhibits peri-infarct neuronal activation as reflected by decreased ECoG gamma power and cFos expression. Meanwhile, lower microglial activation and smaller lesion volumes were observed in animals receiving post-stroke stimulation. Together, these results suggest that acute electrical stimulation can be used safely and effectively as an early stroke intervention to reduce excitotoxicity and inflammation, thus mitigating neural damage and enhancing stroke outcomes.
Collapse
Affiliation(s)
- Jasmine Zhou
- Department of Bioengineering, University of Washington, Seattle, WA, 98195
- Washington National Primate Research Center, Seattle, WA, 98195
| | - Karam Khateeb
- Department of Bioengineering, University of Washington, Seattle, WA, 98195
- Washington National Primate Research Center, Seattle, WA, 98195
| | - Azadeh Yazdan-Shahmorad
- Department of Bioengineering, University of Washington, Seattle, WA, 98195
- Washington National Primate Research Center, Seattle, WA, 98195
- Department of Electrical and Computer Engineering, University of Washington, Seattle, WA, 98195
| |
Collapse
|
53
|
Matur AV, Candelario-Jalil E, Paul S, Karamyan VT, Lee JD, Pennypacker K, Fraser JF. Translating Animal Models of Ischemic Stroke to the Human Condition. Transl Stroke Res 2023; 14:842-853. [PMID: 36125734 DOI: 10.1007/s12975-022-01082-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/30/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022]
Abstract
Ischemic stroke is a leading cause of death and disability. However, very few neuroprotective agents have shown promise for treatment of ischemic stroke in clinical trials, despite showing efficacy in many successful preclinical studies. This may be attributed, at least in part, to the incongruency between experimental animal stroke models used in preclinical studies and the manifestation of ischemic stroke in humans. Most often the human population selected for clinical trials are more diverse than the experimental model used in a preclinical study. For successful translation, it is critical to develop clinical trial designs that match the experimental animal model used in the preclinical study. This review aims to provide a comprehensive summary of commonly used animal models with clear correlates between rodent models used to study ischemic stroke and the clinical stroke pathologies with which they most closely align. By improving the correlation between preclinical studies and clinical trials, new neuroprotective agents and stroke therapies may be more accurately and efficiently identified.
Collapse
Affiliation(s)
- Abhijith V Matur
- Department of Radiology, University of Kentucky, Lexington, KY, USA.
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Surojit Paul
- Department of Neurology and Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Vardan T Karamyan
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI, USA
| | - Jessica D Lee
- Department of Neurology, University of Kentucky, Lexington, KY, USA
| | - Keith Pennypacker
- Department of Neurology, University of Kentucky, Lexington, KY, USA
- Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, USA
| | - Justin F Fraser
- Department of Radiology, University of Kentucky, Lexington, KY, USA
- Department of Neurology, University of Kentucky, Lexington, KY, USA
- Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
- Department of Neurological Surgery, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
54
|
Rogalewski A, Schäbitz W. [Therapies for the Improvement of Stroke Recovery - Assessment of Clinical Trial Results]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2023; 91:516-522. [PMID: 38081165 DOI: 10.1055/a-2181-1026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Recovery processes after stroke include restoration or compensation of function initially lost or newly acquired after injury. Therapeutic interventions can either directly improve these processes and/or inhibit processes that impede regeneration. Numerous experimental studies suggested a great opportunity for such treatments, but the results from recent large clinical trials with neuromodulators such as dopamine and fluoxetine have been rather disappointing. The reasons for this are manifold and involve the extrapolation of results from animal models to humans. Given the differences between animals and humans in genetic and epigenetic background, brain size and anatomy, cerebral vascular anatomy, immune system, as well as clinical function, and behavior, direct extrapolation is unlikely to work. Backward blockades include the incompatible adaption of clinical trial objectives and outcomes in clinical trials with regard to previous preclinical findings. For example, the clinical recovery trial design widely varies and has been characterized by the selection of different clinical endpoints, the inclusion a wide spectrum of stroke subtypes and clinical syndromes, and different time windows for treatment initiation after onset of infarction. This review will discuss these aspects based on the results of the recent stroke recovery trials with the aim to contributing to the development of a therapy that improves the functional outcome of a chronic stroke patient.
Collapse
Affiliation(s)
- Andreas Rogalewski
- Klinik für Neurologie, Sankt Elisabeth-Hospital Gütersloh, Gütersloh, Germany
| | - Wolf Schäbitz
- Universitätsklinik für Neurologie, Evangelisches Klinikum Bethel, Universitätsklinikum OWL der Universität Bielefeld, Campus Bielefeld-Bethel, Bielefeld, Germany
| |
Collapse
|
55
|
Collyer E, Boyle BR, Gomez-Galvez Y, Iacovitti L, Blanco-Suarez E. Absence of chordin-like 1 aids motor recovery in a mouse model of stroke. Exp Neurol 2023; 370:114548. [PMID: 37769794 DOI: 10.1016/j.expneurol.2023.114548] [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: 07/05/2023] [Revised: 09/06/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
Chordin-like 1 (Chrdl1) is an astrocyte-secreted protein that regulates synaptic maturation, and limits plasticity via GluA2-containing AMPA receptors (AMPARs). It was demonstrated that Chrdl1 expression is very heterogeneous throughout the brain, and it is enriched in astrocytes in cortical layers 2/3, with peak expression in the visual cortex at postnatal day 14. In response to ischemic stroke, Chrdl1 is upregulated during the acute and sub-acute phases in the peri-infarct region, potentially hindering recovery after stroke. Here, we used photothrombosis to model ischemic stroke in the motor cortex of adult male and female mice. In this study, we demonstrate that elimination of Chrdl1 in a global knock-out mouse reduces apoptotic cell death at early post-stroke stages and prevents ischemia-driven synaptic loss of AMPA receptors at later time points, all contributing to faster motor recovery. This suggests that synapse-regulating astrocyte-secreted proteins such as Chrdl1 have therapeutic potential to aid functional recovery after an ischemic injury.
Collapse
Affiliation(s)
- Eileen Collyer
- Department of Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Bridget R Boyle
- Department of Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Yolanda Gomez-Galvez
- Department of Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; The Joseph and Marie Field Cerebrovascular Research Laboratory, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lorraine Iacovitti
- Department of Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; Department of Neurological Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; The Joseph and Marie Field Cerebrovascular Research Laboratory, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Elena Blanco-Suarez
- Department of Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; Department of Neurological Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
56
|
Brunner C, Montaldo G, Urban A. Functional ultrasound imaging of stroke in awake rats. eLife 2023; 12:RP88919. [PMID: 37988288 PMCID: PMC10662948 DOI: 10.7554/elife.88919] [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] [Indexed: 11/23/2023] Open
Abstract
Anesthesia is a major confounding factor in preclinical stroke research as stroke rarely occurs in sedated patients. Moreover, anesthesia affects both brain functions and the stroke outcome acting as neurotoxic or protective agents. So far, no approaches were well suited to induce stroke while imaging hemodynamics along with simultaneous large-scale recording of brain functions in awake animals. For this reason, the first critical hours following the stroke insult and associated functional alteration remain poorly understood. Here, we present a strategy to investigate both stroke hemodynamics and stroke-induced functional alterations without the confounding effect of anesthesia, i.e., under awake condition. Functional ultrasound (fUS) imaging was used to continuously monitor variations in cerebral blood volume (CBV) in +65 brain regions/hemispheres for up to 3 hr after stroke onset. The focal cortical ischemia was induced using a chemo-thrombotic agent suited for permanent middle cerebral artery occlusion in awake rats and followed by ipsi- and contralesional whiskers stimulation to investigate on the dynamic of the thalamocortical functions. Early (0-3 hr) and delayed (day 5) fUS recording enabled to characterize the features of the ischemia (location, CBV loss), spreading depolarizations (occurrence, amplitude) and functional alteration of the somatosensory thalamocortical circuits. Post-stroke thalamocortical functions were affected at both early and later time points (0-3 hr and 5 days) after stroke. Overall, our procedure facilitates early, continuous, and chronic assessments of hemodynamics and cerebral functions. When integrated with stroke studies or other pathological analyses, this approach seeks to enhance our comprehension of physiopathologies towards the development of pertinent therapeutic interventions.
Collapse
Affiliation(s)
- Clément Brunner
- Neuro-Electronics Research FlandersLeuvenBelgium
- Vlaams Instituut voor BiotechnologieLeuvenBelgium
- Interuniversity Microelectronics CentreLeuvenBelgium
- Department of Neurosciences, KU LeuvenLeuvenBelgium
| | - Gabriel Montaldo
- Neuro-Electronics Research FlandersLeuvenBelgium
- Vlaams Instituut voor BiotechnologieLeuvenBelgium
- Interuniversity Microelectronics CentreLeuvenBelgium
- Department of Neurosciences, KU LeuvenLeuvenBelgium
| | - Alan Urban
- Neuro-Electronics Research FlandersLeuvenBelgium
- Vlaams Instituut voor BiotechnologieLeuvenBelgium
- Interuniversity Microelectronics CentreLeuvenBelgium
- Department of Neurosciences, KU LeuvenLeuvenBelgium
| |
Collapse
|
57
|
Cuartas-Vélez C, Middelkamp HHT, van der Meer AD, van den Berg A, Bosschaart N. Tracking the dynamics of thrombus formation in a blood vessel-on-chip with visible-light optical coherence tomography. BIOMEDICAL OPTICS EXPRESS 2023; 14:5642-5655. [PMID: 38021142 PMCID: PMC10659801 DOI: 10.1364/boe.500434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/31/2023] [Accepted: 09/03/2023] [Indexed: 12/01/2023]
Abstract
Thrombus formation is a physiological response to damage in a blood vessel that relies on a complex interplay of platelets, coagulation factors, immune cells, and the vessel wall. The dynamics of thrombus formation are essential for a deeper understanding of many disease processes, like bleeding, wound healing, and thrombosis. However, monitoring thrombus formation is challenging due to the limited imaging options available to analyze flowing blood. In this work, we use a visible-light optical coherence tomography (vis-OCT) system to monitor the dynamic process of the formation of thrombi in a microfluidic blood vessel-on-chip (VoC) device. Inside the VoC, thrombi form in a channel lined with a monolayer of endothelial cells and perfused by human whole blood. We show that the correlation of the vis-OCT signal can be utilized as a marker for thrombus formation. By thresholding the correlation during thrombus formation, we track and quantify the growth of the thrombi over time. We validate our results with fluorescence microscopic imaging of fibrin and platelet markers at the end of the blood perfusion assay. In conclusion, we demonstrate that the correlation of the vis-OCT signal can be used to visualize both the spatial and temporal behavior of the thrombus formation in flowing human whole blood.
Collapse
Affiliation(s)
- Carlos Cuartas-Vélez
- Biomedical Photonic Imaging Group, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Heleen H. T. Middelkamp
- BIOS/Lab on a Chip, Faculty of Electrical Engineering, Mathematics and Computer Science, University of Twente, Enschede, The Netherlands
| | - Andries D. van der Meer
- Applied Stem Cell Technologies, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Albert van den Berg
- BIOS/Lab on a Chip, Faculty of Electrical Engineering, Mathematics and Computer Science, University of Twente, Enschede, The Netherlands
| | - Nienke Bosschaart
- Biomedical Photonic Imaging Group, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| |
Collapse
|
58
|
Li Y, Schappell LE, Polizu C, DiPersio J, Tsirka SE, Halterman MW, Nadkarni NA. Evolving Clinical-Translational Investigations of Cerebroprotection in Ischemic Stroke. J Clin Med 2023; 12:6715. [PMID: 37959180 PMCID: PMC10649331 DOI: 10.3390/jcm12216715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/15/2023] Open
Abstract
Ischemic stroke is a highly morbid disease, with over 50% of large vessel stroke (middle cerebral artery or internal carotid artery terminus occlusion) patients suffering disability despite maximal acute reperfusion therapy with thrombolysis and thrombectomy. The discovery of the ischemic penumbra in the 1980s laid the foundation for a salvageable territory in ischemic stroke. Since then, the concept of neuroprotection has been a focus of post-stroke care to (1) minimize the conversion from penumbra to core irreversible infarct, (2) limit secondary damage from ischemia-reperfusion injury, inflammation, and excitotoxicity and (3) to encourage tissue repair. However, despite multiple studies, the preclinical-clinical research enterprise has not yet created an agent that mitigates post-stroke outcomes beyond thrombolysis and mechanical clot retrieval. These translational gaps have not deterred the scientific community as agents are under continuous investigation. The NIH has recently promoted the concept of cerebroprotection to consider the whole brain post-stroke rather than just the neurons. This review will briefly outline the translational science of past, current, and emerging breakthroughs in cerebroprotection and use of these foundational ideas to develop a novel paradigm for optimizing stroke outcomes.
Collapse
Affiliation(s)
- Yinghui Li
- Department of Neurology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794-8651, USA; (Y.L.); (L.E.S.); (C.P.); (J.D.); (M.W.H.)
| | - Laurel E. Schappell
- Department of Neurology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794-8651, USA; (Y.L.); (L.E.S.); (C.P.); (J.D.); (M.W.H.)
- Department of Pharmacological Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794-8651, USA;
| | - Claire Polizu
- Department of Neurology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794-8651, USA; (Y.L.); (L.E.S.); (C.P.); (J.D.); (M.W.H.)
| | - James DiPersio
- Department of Neurology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794-8651, USA; (Y.L.); (L.E.S.); (C.P.); (J.D.); (M.W.H.)
| | - Stella E. Tsirka
- Department of Pharmacological Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794-8651, USA;
| | - Marc W. Halterman
- Department of Neurology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794-8651, USA; (Y.L.); (L.E.S.); (C.P.); (J.D.); (M.W.H.)
| | - Neil A. Nadkarni
- Department of Neurology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794-8651, USA; (Y.L.); (L.E.S.); (C.P.); (J.D.); (M.W.H.)
| |
Collapse
|
59
|
Irie T, Matsuda T, Hayashi Y, Matsuda-Ito K, Kamiya A, Masuda T, Prinz M, Isobe N, Kira JI, Nakashima K. Direct neuronal conversion of microglia/macrophages reinstates neurological function after stroke. Proc Natl Acad Sci U S A 2023; 120:e2307972120. [PMID: 37812721 PMCID: PMC10589698 DOI: 10.1073/pnas.2307972120] [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: 05/16/2023] [Accepted: 09/05/2023] [Indexed: 10/11/2023] Open
Abstract
Although generating new neurons in the ischemic injured brain would be an ideal approach to replenish the lost neurons for repairing the damage, the adult mammalian brain retains only limited neurogenic capability. Here, we show that direct conversion of microglia/macrophages into neurons in the brain has great potential as a therapeutic strategy for ischemic brain injury. After transient middle cerebral artery occlusion in adult mice, microglia/macrophages converge at the lesion core of the striatum, where neuronal loss is prominent. Targeted expression of a neurogenic transcription factor, NeuroD1, in microglia/macrophages in the injured striatum enables their conversion into induced neuronal cells that functionally integrate into the existing neuronal circuits. Furthermore, NeuroD1-mediated induced neuronal cell generation significantly improves neurological function in the mouse stroke model, and ablation of these cells abolishes the gained functional recovery. Our findings thus demonstrate that neuronal conversion contributes directly to functional recovery after stroke.
Collapse
Affiliation(s)
- Takashi Irie
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 812-8582Fukuoka, Japan
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 812-8582Fukuoka, Japan
| | - Taito Matsuda
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 812-8582Fukuoka, Japan
| | - Yoshinori Hayashi
- Department of Physiology, Nihon University School of Dentistry, 101-8310Tokyo, Japan
| | - Kanae Matsuda-Ito
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 812-8582Fukuoka, Japan
| | - Akihide Kamiya
- Department of Molecular Life Sciences, Tokai University School of Medicine, 259-1193Isehara, Japan
- Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 108-8639Tokyo, Japan
| | - Takahiro Masuda
- Division of Molecular Neuroinflammation, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, 812-8582Fukuoka, Japan
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, D-79106Freiburg, Germany
- Signalling Research Centres Centre for Biological Signalling Studies and Centre for Integrative Biological Signalling Studies, University of Freiburg, D-79106Freiburg, Germany
| | - Noriko Isobe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 812-8582Fukuoka, Japan
| | - Jun-ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 812-8582Fukuoka, Japan
- Translational Neuroscience Center, Graduate School of Medicine, and School of Pharmacy at Fukuoka, International University of Health and Welfare, 831-8501Okawa, Japan
- Department of Neurology, Brain and Nerve Center, Fukuoka Central Hospital, International University of Health and Welfare, 810-0022Fukuoka, Japan
| | - Kinichi Nakashima
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 812-8582Fukuoka, Japan
| |
Collapse
|
60
|
Yan Q, Yin Y, Li X, Li M. Exosome-shuttled MYCBPAP from bone marrow mesenchymal stem cells regulates synaptic remodeling and ameliorates ischemic stroke in rats. J Chem Neuroanat 2023; 132:102309. [PMID: 37423468 DOI: 10.1016/j.jchemneu.2023.102309] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/19/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND AND PURPOSE Mesenchymal stem cells (MSC) have been demonstrated to improve cardiac function via the secretion of paracrine factors rather than direct differentiation. We, therefore, investigated whether bone marrow-derived MSC (BMSC)-released exosomes (BMSC-exo) enhance neurological recovery in spontaneously hypertensive rats (SHR) with ischemic stroke. METHODS Markers of BMSC and BMSC-exo were detected to characterize BMSC and BMSC-exo. A green fluorescent PKH-67-labeled assay was conducted to ensure BMSC-exo internalization. Rat neuronal cells (RNC) were induced with Ang II and oxygen-glucose deprivation. The protective effects of BMSC-exo on RNC were studied by CCK-8, LDH, and immunofluorescence assays. SHR were subjected to middle cerebral artery occlusion, and the systolic and diastolic blood pressure changes in the modeled rats were measured. The effects of BMSC-exo on SHR were investigated by mNSS scoring, foot-fault tests, immunohistochemistry, Western blot, TTC staining, TUNEL, and HE staining. The hub genes related to SHR and proteins shuttled by BMSC-exo were intersected to obtain a possible candidate, followed by rescue experiments. RESULTS BMSC-exo significantly promoted RNC viability and repressed cell apoptosis and cytotoxicity. Moreover, SHR administrated with BMSC-exo exhibited significant improvement in functional recovery and narrowed infarct size. BMSC-exo shuttled the MYCBPAP protein. Knockdown of MYCBPAP inhibited the protective effects of BMSC-exo on RNC and exacerbated synaptic damage in SHR. CONCLUSIONS MYCBPAP shuttled by BMSC-exo facilitates synaptic remodeling in SHR, which may contribute to a therapeutic strategy for ischemic stroke treatment.
Collapse
Affiliation(s)
- Qiuyue Yan
- Department of Neurology, Cangzhou Central Hospital, Cangzhou 061001 Hebei, PR China.
| | - Yong Yin
- Department of Neurology, Cangzhou Central Hospital, Cangzhou 061001 Hebei, PR China
| | - Xuechun Li
- Department of Neurology, Cangzhou Central Hospital, Cangzhou 061001 Hebei, PR China
| | - Meng Li
- Department of Neurology, Cangzhou Central Hospital, Cangzhou 061001 Hebei, PR China
| |
Collapse
|
61
|
Wu X, Li JR, Fu Y, Chen DY, Nie H, Tang ZP. From static to dynamic: live observation of the support system after ischemic stroke by two photon-excited fluorescence laser-scanning microscopy. Neural Regen Res 2023; 18:2093-2107. [PMID: 37056116 PMCID: PMC10328295 DOI: 10.4103/1673-5374.369099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/21/2022] [Accepted: 01/13/2023] [Indexed: 02/17/2023] Open
Abstract
Ischemic stroke is one of the most common causes of mortality and disability worldwide. However, treatment efficacy and the progress of research remain unsatisfactory. As the critical support system and essential components in neurovascular units, glial cells and blood vessels (including the blood-brain barrier) together maintain an optimal microenvironment for neuronal function. They provide nutrients, regulate neuronal excitability, and prevent harmful substances from entering brain tissue. The highly dynamic networks of this support system play an essential role in ischemic stroke through processes including brain homeostasis, supporting neuronal function, and reacting to injuries. However, most studies have focused on postmortem animals, which inevitably lack critical information about the dynamic changes that occur after ischemic stroke. Therefore, a high-precision technique for research in living animals is urgently needed. Two-photon fluorescence laser-scanning microscopy is a powerful imaging technique that can facilitate live imaging at high spatiotemporal resolutions. Two-photon fluorescence laser-scanning microscopy can provide images of the whole-cortex vascular 3D structure, information on multicellular component interactions, and provide images of structure and function in the cranial window. This technique shifts the existing research paradigm from static to dynamic, from flat to stereoscopic, and from single-cell function to multicellular intercommunication, thus providing direct and reliable evidence to identify the pathophysiological mechanisms following ischemic stroke in an intact brain. In this review, we discuss exciting findings from research on the support system after ischemic stroke using two-photon fluorescence laser-scanning microscopy, highlighting the importance of dynamic observations of cellular behavior and interactions in the networks of the brain's support systems. We show the excellent application prospects and advantages of two-photon fluorescence laser-scanning microscopy and predict future research developments and directions in the study of ischemic stroke.
Collapse
Affiliation(s)
- Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jia-Rui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yu Fu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Dan-Yang Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hao Nie
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhou-Ping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
62
|
Collyer E, Blanco-Suarez E. Astrocytes in stroke-induced neurodegeneration: a timeline. FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1240862. [PMID: 39086680 PMCID: PMC11285566 DOI: 10.3389/fmmed.2023.1240862] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/22/2023] [Indexed: 08/02/2024]
Abstract
Stroke is a condition characterized by sudden deprivation of blood flow to a brain region and defined by different post-injury phases, which involve various molecular and cellular cascades. At an early stage during the acute phase, fast initial cell death occurs, followed by inflammation and scarring. This is followed by a sub-acute or recovery phase when endogenous plasticity mechanisms may promote spontaneous recovery, depending on various factors that are yet to be completely understood. At later time points, stroke leads to greater neurodegeneration compared to healthy controls in both clinical and preclinical studies, this is evident during the chronic phase when recovery slows down and neurodegenerative signatures appear. Astrocytes have been studied in the context of ischemic stroke due to their role in glutamate re-uptake, as components of the neurovascular unit, as building blocks of the glial scar, and synaptic plasticity regulators. All these roles render astrocytes interesting, yet understudied players in the context of stroke-induced neurodegeneration. With this review, we provide a summary of previous research, highlight astrocytes as potential therapeutic targets, and formulate questions about the role of astrocytes in the mechanisms during the acute, sub-acute, and chronic post-stroke phases that may lead to neurorestoration or neurodegeneration.
Collapse
Affiliation(s)
| | - Elena Blanco-Suarez
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
63
|
Pinky, Neha, Salman M, Kumar P, Khan MA, Jamal A, Parvez S. Age-related pathophysiological alterations in molecular stress markers and key modulators of hypoxia. Ageing Res Rev 2023; 90:102022. [PMID: 37490963 DOI: 10.1016/j.arr.2023.102022] [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: 04/09/2023] [Revised: 06/30/2023] [Accepted: 07/21/2023] [Indexed: 07/27/2023]
Abstract
Alzheimer's disease (AD) is characterized by an adverse cellular environment and pathological alterations in distinct brain regions. The development is triggered or facilitated by a condition such as hypoxia or ischemia, or inflammation and is associated with disruptions of fundamental cellular functions, including metabolic and ion homeostasis. Increasing evidence suggests that hypoxia may affect many pathological aspects of AD, including oxidative stress, mitochondrial dysfunction, ER stress, amyloidogenic processing of APP, and Aβ accumulation, which may collectively result in neurodegeneration. Further investigation into the relationship between hypoxia and AD may provide an avenue for the effective preservation and pharmacological treatment of this neurodegenerative disease. This review summarizes the effects of normoxia and hypoxia on AD pathogenesis and discusses the underlying mechanisms. Regulation of HIF-1α and the role of its key players, including P53, VEGF, and GLUT1, are also discussed.
Collapse
Affiliation(s)
- Pinky
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| | - Neha
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| | - Mohd Salman
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| | - Pratika Kumar
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| | - Mohammad Ahmed Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.
| | - Azfar Jamal
- Department of Biology, College of Science, Al-Zulfi-, Majmaah University, Al-Majmaah 11952, Saudi Arabia; Health and Basic Science Research Centre, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
64
|
Dzirkale Z, Pilipenko V, Pijet B, Klimaviciusa L, Upite J, Protokowicz K, Kaczmarek L, Jansone B. Long-term behavioural alterations in mice following transient cerebral ischemia. Behav Brain Res 2023; 452:114589. [PMID: 37481076 DOI: 10.1016/j.bbr.2023.114589] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/05/2023] [Accepted: 07/17/2023] [Indexed: 07/24/2023]
Abstract
Ischemic stroke is one of the leading causes of disability and mortality worldwide. Acute and chronic post-stroke changes have variable effects on the functional outcomes of the disease. Therefore, it is imperative to identify what daily activities are altered after stroke and to what extent, keeping in mind that ischemic stroke patients often have long-term post-stroke complications. Translational studies in stroke have also been challenging due to inconsistent study design of animal experiments. The objective of this study was to clarify whether and to what extent mouse behaviour was altered during a 6 months period after cerebral stroke. Experimental stroke was induced in mice by intraluminal filament insertion into the middle cerebral artery (fMCAo). Neurological deficits, recovery rate, motor performance, and circadian activity were evaluated following ischemia. We observed severe neurological deficits, motor impairments, and delay in the recovery rate of mice during the first 14 days after fMCAo. Aberrant circadian activity and distorted space map were seen in fMCAo mice starting one month after ischemia, similarly to altered new and familiar cage activity and sucrose preference using the IntelliCage, and was still evident 60- and 180- days following stroke in the voluntary running wheel using the PhenoMaster system. A preference towards ipsilateral side turns was observed in fMCAo mice both acutely and chronically after the stroke induction. Overall, our study shows the importance of determining time-dependent differences in the long-term post-stroke recovery (over 180 days after fMCAo) using multiple behavioural assessments.
Collapse
Affiliation(s)
- Zane Dzirkale
- Department of Pharmacology, Faculty of Medicine, University of Latvia, 3 Jelgavas Street, LV-1004 Riga, Latvia.
| | - Vladimirs Pilipenko
- Department of Pharmacology, Faculty of Medicine, University of Latvia, 3 Jelgavas Street, LV-1004 Riga, Latvia
| | - Barbara Pijet
- Laboratory of Neurobiology, BRAINCITY - Centre of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Linda Klimaviciusa
- Department of Pharmacology, Faculty of Medicine, University of Latvia, 3 Jelgavas Street, LV-1004 Riga, Latvia
| | - Jolanta Upite
- Department of Pharmacology, Faculty of Medicine, University of Latvia, 3 Jelgavas Street, LV-1004 Riga, Latvia
| | - Karolina Protokowicz
- Laboratory of Neurobiology, BRAINCITY - Centre of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, BRAINCITY - Centre of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Baiba Jansone
- Department of Pharmacology, Faculty of Medicine, University of Latvia, 3 Jelgavas Street, LV-1004 Riga, Latvia.
| |
Collapse
|
65
|
Jacques MT, de Souza V, Barbosa FAR, Faria Santos Canto R, Lopes SC, Prediger RD, Braga AL, Aschner M, Farina M. Novel Probucol Analogue, 4,4'-Diselanediylbis (2,6-Di- tert-Butylphenol), Prevents Oxidative Glutamate Neurotoxicity In Vitro and Confers Neuroprotection in a Rodent Model of Ischemic Stroke. ACS Chem Neurosci 2023; 14:2857-2867. [PMID: 37499207 DOI: 10.1021/acschemneuro.3c00138] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023] Open
Abstract
Oxidative glutamate toxicity is regarded as one of the injurious mechanisms associated with ischemic stroke, which represents a major health problem and requires improved pharmacological treatments. We designed and synthesized two new probucol analogues [2,6-di-tert-butyl-4-selenocyanatophenol (C1) and 4,4'-diselanediylbis (2,6-di-tert-butylphenol) (C2)] and investigated their effects against glutamate-induced neuronal oxidative toxicity in vitro in cultured HT22 cells, compared with their parental compound (probucol). In addition, C2, which exhibited the lowest toxicity, was investigated in an in vivo rodent model of ischemic stroke. Glutamate caused concentration- and time-dependent cytotoxicity in HT22 neuronal cells, which was preceded by increased levels of oxidants and depletion of the antioxidant glutathione. The analogues (C1 and C2), but not probucol, significantly decreased the levels of oxidants (including mitochondrial superoxide anion and lipid reactive oxygen species (ROS)) and protected against glutamate-induced cytotoxicity. In the in vivo model of ischemic stroke, which was based on central injections of the vasoconstrictor agent endothelin-1 (800 pmol/site), C2 (20 or 50 mg/kg/day, intraperitoneally, for 4 consecutive days after stroke) displayed significant beneficial effects against ischemic injury in vivo, improving rats' motor-related behavioral skills and decreasing stroke-related striatal gliosis. This is the first study to design, synthesize, and present a probucol analogue (C2) with in vivo beneficial effects against ischemic stroke. This novel compound, which was able to mitigate glutamate-induced oxidative toxicity in vitro, represents a promising neuroprotective drug.
Collapse
Affiliation(s)
- Mauricio Tavares Jacques
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Viviane de Souza
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | | | - Rômulo Faria Santos Canto
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, RS, Brazil
| | - Samantha Cristiane Lopes
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Rui Daniel Prediger
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Antônio Luiz Braga
- Department of Chemistry, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461, United States
| | - Marcelo Farina
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| |
Collapse
|
66
|
Modi JP, Shen W, Menzie-Suderam J, Xu H, Lin CH, Tao R, Prentice HM, Schloss J, Wu JY. The Role of NMDA Receptor Partial Antagonist, Carbamathione, as a Therapeutic Agent for Transient Global Ischemia. Biomedicines 2023; 11:1885. [PMID: 37509524 PMCID: PMC10377037 DOI: 10.3390/biomedicines11071885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Carbamathione (Carb), an NMDA glutamate receptor partial antagonist, has potent neuroprotective functions against hypoxia- or ischemia-induced neuronal injury in cell- or animal-based stroke models. We used PC-12 cell cultures as a cell-based model and bilateral carotid artery occlusion (BCAO) for stroke. Whole-cell patch clamp recording in the mouse retinal ganglion cells was performed. Key proteins involved in apoptosis, endoplasmic reticulum (ER) stress, and heat shock proteins were analyzed using immunoblotting. Carb is effective in protecting PC12 cells against glutamate- or hypoxia-induced cell injury. Electrophysiological results show that Carb attenuates NMDA-mediated glutamate currents in the retinal ganglion cells, which results in activation of the AKT signaling pathway and increased expression of pro-cell survival biomarkers, e.g., Hsp 27, P-AKT, and Bcl2 and decreased expression of pro-cell death markers, e.g., Beclin 1, Bax, and Cleaved caspase 3, and ER stress markers, e.g., CHOP, IRE1, XBP1, ATF 4, and eIF2α. Using the BCAO animal stroke model, we found that Carb reduced the brain infarct volume and decreased levels of ER stress markers, GRP 78, CHOP, and at the behavioral level, e.g., a decrease in asymmetric turns and an increase in locomotor activity. These findings for Carb provide promising and rational strategies for stroke therapy.
Collapse
Affiliation(s)
- Jigar Pravinchandra Modi
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
- Center of Complex Systems and Brain Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Wen Shen
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
- Program in Integrative Biology, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Janet Menzie-Suderam
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
- Program in Integrative Biology, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Hongyuan Xu
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Chun-Hua Lin
- Department of Nursing, Kang-Ning University, Taipei 11485, Taiwan
| | - Rui Tao
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
- Program in Integrative Biology, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Howard M Prentice
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
- Center of Complex Systems and Brain Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
- Program in Integrative Biology, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - John Schloss
- Department of Pharmaceutical Science, American University of Health Sciences, Signal Hill, CA 90755, USA
| | - Jang-Yen Wu
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
- Center of Complex Systems and Brain Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
- Program in Integrative Biology, Florida Atlantic University, Boca Raton, FL 33431, USA
| |
Collapse
|
67
|
Zeng L, Hu S, Zeng L, Chen R, Li H, Yu J, Yang H. Animal Models of Ischemic Stroke with Different Forms of Middle Cerebral Artery Occlusion. Brain Sci 2023; 13:1007. [PMID: 37508939 PMCID: PMC10377124 DOI: 10.3390/brainsci13071007] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Ischemic stroke is a common type of stroke that significantly affects human well-being and quality of life. In order to further characterize the pathophysiology of ischemic stroke and develop new treatment strategies, ischemic stroke models with controllable and consistent response to potential clinical treatments are urgently needed. The middle cerebral artery occlusion (MCAO) model is currently the most widely used animal model of ischemic stroke. This review discusses various methods for constructing the MCAO model and compares their advantages and disadvantages in order to provide better approaches for studying ischemic stroke.
Collapse
Affiliation(s)
- Lang Zeng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shengqi Hu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lingcheng Zeng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Rudong Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hua Li
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jiasheng Yu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hongkuan Yang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
68
|
Li Y, Tan L, Yang C, He L, Liu L, Deng B, Liu S, Guo J. Distinctions between the Koizumi and Zea Longa methods for middle cerebral artery occlusion (MCAO) model: a systematic review and meta-analysis of rodent data. Sci Rep 2023; 13:10247. [PMID: 37353569 PMCID: PMC10290095 DOI: 10.1038/s41598-023-37187-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/17/2023] [Indexed: 06/25/2023] Open
Abstract
Ischemic stroke in rodents is usually induced by intraluminal middle cerebral artery occlusion (MCAO) via the common carotid artery plugging filament invented by Koizumi et al. (MCAO-KM), or the external carotid artery plugging filament created by Zea Longa et al. (MCAO-LG). A systematic review of the distinctions between them is currently lacking. Here, we performed a meta-analysis in terms of model establishment, cerebral blood flow (CBF), and cerebral ischemia-reperfusion injury (CIRI) between them, Weighted Mean Differences and Standardized Mean Difference were used to analyze the combined effects, Cochrane's Q test and the I2 statistic were applied to determine heterogeneity, sensitivity analysis and subgroup analysis were performed to explore the source of heterogeneity. Literature mining suggests that MCAO-KM brings shorter operation time (p = 0.007), higher probability of plugging filament (p < 0.001) and molding establishment (p = 0.006), lower possibility of subarachnoid hemorrhage (p = 0.02), larger infarct volume (p = 0.003), severer brain edema (p = 0.002), and neurological deficits (p = 0.03). Nevertheless, MCAO-LG shows a more adequate CBF after ischemia-reperfusion (p < 0.001), a higher model survival rate (p = 0.02), and a greater infarct rate (p = 0.007). In conclusion, the MCAO-KM method is simple to operate with a high modeling success rate, and is suitable for the study of brain edema under long-term hypoperfusion, while the MCAO-LG method is highly challenging for novices, and is suitable for the study of CIRI caused by complete ischemia-reperfusion. These findings are expected to benefit the selection of intraluminal filament MCAO models before undertaking ischemic stroke preclinical effectiveness trials.
Collapse
Affiliation(s)
- Yong Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Tan
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Caixia Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liying He
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin Liu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bowen Deng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sijing Liu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinlin Guo
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
69
|
Rust R. Ischemic stroke-related gene expression profiles across species: a meta-analysis. J Inflamm (Lond) 2023; 20:21. [PMID: 37337154 DOI: 10.1186/s12950-023-00346-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/24/2023] [Indexed: 06/21/2023] Open
Abstract
Stroke patients are often left with permanent disabilities with no regenerative treatment options. Unbiased RNA sequencing studies decoding the transcriptional signature of stroked tissue hold promise to identify new potential targets and pathways directed to improve treatment for stroke patients. Here, gene expression profiles of stroked tissue across different time points, species, and stroke models were compared using NCBI GEO database. In total, 34 datasets from mice, rats, humans, and primates were included, exploring gene expression differences in healthy and stroked brain tissue. Distinct changes in gene expression and pathway enrichment revealed the heterogenicity of the stroke pathology in stroke-related pathways e.g., inflammatory responses, vascular repair, remodelling and cell proliferation and adhesion but also in diverse general, stroke-unrelated pathways that have to be carefully considered when evaluating new promising therapeutic targets.
Collapse
Affiliation(s)
- Ruslan Rust
- Institute for Regenerative Medicine (IREM), University of Zurich, Campus Schlieren Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland.
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
70
|
Miura T, Kouno T, Takano M, Kuroda T, Yamamoto Y, Kusakawa S, Morioka MS, Sugawara T, Hirai T, Yasuda S, Sawada R, Matsuyama S, Kawaji H, Kasukawa T, Itoh M, Matsuyama A, Shin JW, Umezawa A, Kawai J, Sato Y. Single-Cell RNA-Seq Reveals LRRC75A-Expressing Cell Population Involved in VEGF Secretion of Multipotent Mesenchymal Stromal/Stem Cells Under Ischemia. Stem Cells Transl Med 2023; 12:379-390. [PMID: 37263619 PMCID: PMC10267575 DOI: 10.1093/stcltm/szad029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 04/15/2023] [Indexed: 06/03/2023] Open
Abstract
Human multipotent mesenchymal stromal/stem cells (MSCs) have been utilized in cell therapy for various diseases and their clinical applications are expected to increase in the future. However, the variation in MSC-based product quality due to the MSC heterogeneity has resulted in significant constraints in the clinical utility of MSCs. Therefore, we hypothesized that it might be important to identify and ensure/enrich suitable cell subpopulations for therapies using MSC-based products. In this study, we aimed to identify functional cell subpopulations to predict the efficacy of angiogenic therapy using bone marrow-derived MSCs (BM-MSCs). To assess its angiogenic potency, we observed various levels of vascular endothelial growth factor (VEGF) secretion among 11 donor-derived BM-MSC lines under in vitro ischemic culture conditions. Next, by clarifying the heterogeneity of BM-MSCs using single-cell RNA-sequencing analysis, we identified a functional cell subpopulation that contributed to the overall VEGF production in BM-MSC lines under ischemic conditions. We also found that leucine-rich repeat-containing 75A (LRRC75A) was more highly expressed in this cell subpopulation than in the others. Importantly, knockdown of LRRC75A using small interfering RNA resulted in significant inhibition of VEGF secretion in ischemic BM-MSCs, indicating that LRRC75A regulates VEGF secretion under ischemic conditions. Therefore, LRRC75A may be a useful biomarker to identify cell subpopulations that contribute to the angiogenic effects of BM-MSCs. Our work provides evidence that a strategy based on single-cell transcriptome profiles is effective for identifying functional cell subpopulations in heterogeneous MSC-based products.
Collapse
Affiliation(s)
- Takumi Miura
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Tsukasa Kouno
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Megumi Takano
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
| | - Takuya Kuroda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
| | - Yumiko Yamamoto
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Shinji Kusakawa
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
| | | | - Tohru Sugawara
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
- Biopharmaceutical and Regenerative Sciences, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Takamasa Hirai
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
| | - Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
| | - Rumi Sawada
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
| | - Satoko Matsuyama
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
- Center for Reverse TR, Osaka Habikino Medical Center, Osaka Prefectural Hospital Organization, Osaka, Japan
| | - Hideya Kawaji
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Research Center for Genome & Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takeya Kasukawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Masayoshi Itoh
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Akifumi Matsuyama
- Center for Reverse TR, Osaka Habikino Medical Center, Osaka Prefectural Hospital Organization, Osaka, Japan
| | - Jay W Shin
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Genomic Institute of Singapore, Agency for Science, Technology and Research, Singapore
| | - Akihiro Umezawa
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Jun Kawai
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Life Science Technology Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
- Life Science Technology Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan
- Department of Cellular and Gene Therapy Products, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| |
Collapse
|
71
|
Prehn A, Hobusch C, Härtig W, Michalski D, Krueger M, Flachmeyer B. Increasing reproducibility in preclinical stroke research: the correlation of immunofluorescence intensity measurements and Western blot analyses strongly depends on antibody clonality and tissue pre-treatment in a mouse model of focal cerebral ischemia. Front Cell Neurosci 2023; 17:1183232. [PMID: 37342767 PMCID: PMC10277931 DOI: 10.3389/fncel.2023.1183232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/16/2023] [Indexed: 06/23/2023] Open
Abstract
In the setting of stroke, ischemia not only impairs neuronal function, but also detrimentally affects the different components of the neurovascular unit, which are shown to be involved in the transition from reversible to long-lasting tissue damage. In this context, the glial proteins myelin basic protein (MBP) and the 2',3'-cyclic-nucleotide 3'-phosphodiesterase (CNP) as well as the vasculature-associated basement membrane proteins laminin and collagen IV have been identified as ischemia-sensitive elements. However, available data from immunofluorescence and Western blot analyses are often found to be contradictory, which renders interpretation of the respective data rather difficult. Therefore, the present study investigates the impact of tissue pre-treatment and antibody clonality on immunofluorescence measurements of the mentioned proteins in a highly reproducible model of permanent middle cerebral artery occlusion. Here, immunofluorescence labeling using polyclonal antibodies revealed an increased immunofluorescence intensity of MBP, CNP, laminin and collagen IV in ischemic areas, although Western blot analyses did not reveal increased protein levels. Importantly, contrary to polyclonal antibodies, monoclonal ones did not provide increased fluorescence intensities in ischemic areas. Further, we were able to demonstrate that different ways of tissue pre-treatment including paraformaldehyde fixation and antigen retrieval may not only impact on fluorescence intensity measurements in general, but rather one-sidedly affect either ischemic or unaffected tissue. Therefore, immunofluorescence intensity measurements do not necessarily correlate with the actual protein levels, especially in ischemia-affected tissue and should always be complemented by different techniques to enhance reproducibility and to hopefully overcome the translational roadblock from bench to bedside.
Collapse
Affiliation(s)
- Anna Prehn
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | | | - Wolfgang Härtig
- Paul Flechsig Institute of Brain Research, Leipzig University, Leipzig, Germany
| | | | - Martin Krueger
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | | |
Collapse
|
72
|
Hakobyan N, Ilerhunmwuwa N, Wasifuddin M, Jamal F, Zagoruychenko T. An Unusual Source of Cerebral Embolism Caused by Lambl's Excrescences. Cureus 2023; 15:e40910. [PMID: 37496536 PMCID: PMC10366472 DOI: 10.7759/cureus.40910] [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: 06/24/2023] [Indexed: 07/28/2023] Open
Abstract
Lambl's excrescences (LEs) are delicate filiform strands formed by connective tissue located along the valve closure lines within the cardiovascular system. Most cases of these excrescences manifest without discernible symptoms, and the exact etiological factors contributing to their formation remain unknown. These excrescences may embolize to the brain, causing strokes. It is essential that all other possible causes of stroke be eliminated prior to identifying Lambl's excrescences as the cause of the stroke. Herein, we present a case of a patient who suffered a stroke, and all conventional testing for common causes of embolic strokes was ruled out. In pursuit of a comprehensive evaluation encompassing the classification of the stroke as cryptogenic, a transesophageal echocardiogram (TEE) was performed, which effectively disclosed the existence of LEs situated on the aortic valve leaflets. The patient was treated with anticoagulation and discharged with close follow-up monitoring. To culminate, the inclusion of this case within our study augments the currently scarce pool of instances that exhibit such characteristic cardioembolic phenomena, thereby accentuating the necessity for additional prospective investigations to substantiate the existence of a causative association linking LEs and cardioembolic strokes.
Collapse
Affiliation(s)
- Narek Hakobyan
- Internal Medicine, Brookdale University Hospital Medical Center, New York City, USA
| | | | - Mustafa Wasifuddin
- Internal Medicine, Brookdale University Hospital Medical Center, New York City, USA
| | - Fares Jamal
- Internal Medicine, Brookdale University Hospital Medical Center, New York City, USA
| | | |
Collapse
|
73
|
Tang Z, Meng S, Song Z, Yang X, Li X, Guo H, Du M, Chen J, Zhu YZ, Wang X. Neutrophil membrane fusogenic nanoliposomal leonurine for targeted ischemic stroke therapy via remodeling cerebral niche and restoring blood-brain barrier integrity. Mater Today Bio 2023; 20:100674. [PMID: 37273794 PMCID: PMC10238753 DOI: 10.1016/j.mtbio.2023.100674] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/19/2023] [Accepted: 05/18/2023] [Indexed: 06/06/2023] Open
Abstract
Ischemic stroke (IS) constitutes the leading cause of global morbidity and mortality. Neuroprotectants are essential to ameliorate the clinical prognosis, but their therapeutic outcomes are tremendously compromised by insufficient delivery to the ischemic lesion and intricate pathogenesis associated with neuronal damage, oxidative stress, inflammation responses, blood-brain barrier (BBB) dysfunction, etc. Herein, a biomimetic nanosystem (Leo@NM-Lipo) composed of neutrophil membrane-fused nanoliposomal leonurine (Leo) is constructed, which can not only efficiently penetrate and repair the disrupted BBB but also robustly remodel the harsh cerebral microenvironment to reverse ischemia-reperfusion (I/R) injury. More specifically, the neutrophil membrane inherits the BBB penetrating, infarct core targeting, inflammation neutralization, and immune evasion properties of neutrophils, while Leo, a naturally occurring neuroprotectant, exerts pleiotropic effects to attenuate brain damage. Remarkably, comprehensive investigations disclose the critical factors influencing the targetability and therapeutic performances of biomimetic nanosystems. Leo@NM-Lipo with a low membrane protein-to-lipid ratio of 1:10 efficiently targets the ischemic lesion and rescues the injured brain by alleviating neuronal apoptosis, oxidative stress, neuroinflammation, and restoring BBB integrity in transient middle cerebral artery occlusion (tMCAO) rats. Taken together, our study provides a neutrophil-mimetic nanoplatform for targeted IS therapy and sheds light on the rational design of biomimetic nanosystems favoring wide medical applications.
Collapse
Affiliation(s)
- Zhuang Tang
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Shiyu Meng
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Zhiling Song
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Xiaoxue Yang
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Xinzhi Li
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Hui Guo
- School of Chemical Engineering and Technology, Sun Yat-sen University, Zhuhai 519082, China
| | - Meirong Du
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Jun Chen
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yi Zhun Zhu
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Xiaolin Wang
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| |
Collapse
|
74
|
Zhang Y, Zhong C, Wang Q, Zhang J, Zhao H, Huang Y, Zhao D, Yang J. Nanoemulsions of Hydroxysafflor Yellow A for Enhancing Physicochemical and In Vivo Performance. Int J Mol Sci 2023; 24:ijms24108658. [PMID: 37240000 DOI: 10.3390/ijms24108658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/13/2023] [Accepted: 04/16/2023] [Indexed: 05/28/2023] Open
Abstract
Stroke was always a disease that threatened human life and health worldwide. We reported the synthesis of a new type of hyaluronic acid-modified multi-walled carbon nanotube. Then, we produced hydroxysafflor yellow A-hydroxypropyl-β-cyclodextrin phospholipid complex water-in-oil nanoemulsion with hyaluronic acid-modified multi-walled carbon nanotubes and chitosan (HC@HMC) for oral treatment of an ischemic stroke. We measured the intestinal absorption and pharmacokinetics of HC@HMC in rats. We found that the intestinal absorption and the pharmacokinetic behavior of HC@HMC was superior to that of HYA. We measured intracerebral concentrations after oral administration of HC@HMC and found that more HYA crossed the blood-brain barrier (BBB) in mice. Finally, we evaluated the efficacy of HC@HMC in middle cerebral artery occlusion/reperfusion (MCAO/R)-injured mice. In MCAO/R mice, oral administration of HC@HMC demonstrated significant protection against cerebral ischemia-reperfusion injury (CIRI). Furthermore, we found HC@HMC may exert a protective effect on cerebral ischemia-reperfusion injury through the COX2/PGD2/DPs pathway. These results suggest that oral administration of HC@HMC may be a potential therapeutic strategy for the treatment of stroke.
Collapse
Affiliation(s)
- Yingjie Zhang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing 400016, China
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Cailing Zhong
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing 400016, China
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Qiong Wang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing 400016, China
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Jingqing Zhang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing 400016, China
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Hua Zhao
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Yuru Huang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Dezhang Zhao
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Junqing Yang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
75
|
Zhao Y, Wang Y, Geng D, Wang Y. Editorial: Neuroinflammation and neuroimmune response in experimental MCAO and ischemic stroke. Front Aging Neurosci 2023; 15:1195395. [PMID: 37251799 PMCID: PMC10213931 DOI: 10.3389/fnagi.2023.1195395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 04/17/2023] [Indexed: 05/31/2023] Open
Affiliation(s)
- Yingzhe Zhao
- Department of Neurology II, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Yuge Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yanqiang Wang
- Department of Neurology II, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
76
|
Yu XJ, Zhang T, Wei ZZ, Gu B, Guo T, Jiang WJ, Shen YQ, Wang D, Wang Q, Wang J. Abnormal expression of miRNA-122 in cerebral infarction and related mechanism of regulating vascular endothelial cell proliferation and apoptosis by targeting CCNG1. Clinics (Sao Paulo) 2023; 78:100199. [PMID: 37119591 PMCID: PMC10173405 DOI: 10.1016/j.clinsp.2023.100199] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 02/21/2023] [Accepted: 03/08/2023] [Indexed: 05/01/2023] Open
Abstract
OBJECTIVE To analyze the value of serum miRNA-122 expression in the diagnosis, severity, and prognosis of Acute Cerebral Infarction (ACI) and the correlation mechanism of serum miRNA-122 on the proliferation and apoptosis of vascular endothelial cells in ACI. METHOD A total of 60 patients with ACI who were admitted to the emergency department of the Taizhou People's Hospital from January 1, 2019, to December 30, 2019, and 30 healthy controls during the same period were selected. General clinical data of all patients at admission were collected. Including age, sex, medical history, and inflammatory factors (C-Reactive Protein [CRP], Interleukin-6 [IL-6], Procalcitonin [PCT], Neutrophil Gelatinase-Associated Lipid carrier protein [NGAL]). The National Institutes of Health Stroke Scale (NIHSS) score at admission and short-term prognosis (the Modified Rankin Score [mRS]) score at 3 months after onset were recorded. The expression level of miRNA-122 in the serum of patients with ACI and normal controls was detected by reverse-transcription quantitative Real-Time Polymerase Chain Reaction (RT-QPCR), and the correlation between the expression level of miRNA-122 in the serum of patients with ACI and the level of inflammatory factors, NIHSS and mRS scores were analyzed. The expression levels of miRNA-122 in the serum of patients with ACI, normal people, and Human Umbilical cord Endothelial Cells (HUVECs) cultured in a blank control group were detected by RT-QPCR and statistically analyzed. MTT and flow cytometry was used to compare the proliferation and apoptosis of vascular endothelial cells in the miRNA-122 mimics and inhibitors transfection groups and the corresponding negative control group. The mRNA and protein levels of apoptosis-related factors Bax, Bcl-2, Caspase-3, and angiogenesis-related proteins Hes1, Notch1, Vascular Endothelial Growth Factors (VEGF), and CCNG1 were detected by RT-QPCR and Western blot. Bioinformatics methods predicted CCNG1 to be the target of miRNA-122, and the direct targeting relationship between CCNG1 and miRNA-122 was verified by a dual-luciferase reporting assay. RESULT Serum miRNA-122 expression in patients with ACI was significantly higher than that in healthy controls, with an area under the receiver operating characteristic curve of 0.929, 95% Confidence Interval of 0.875‒0.983, and an optimal cut-off value of 1.397. The expression levels of CRP, IL-6, and NGAL in patients with ACI were higher than those in healthy control groups, p < 0.05; miRNA-122 was positively correlated with CPR, IL-6, NIHSS score, and mRS score. At 48h and 72h, the proliferation rate of HUVECs cells in the miRNA-122 mimics group decreased and the apoptosis rate increased. Cell proliferation rate increased, and apoptosis rate decreased significantly in the groups transfected with miRNA-122 inhibitors. The mRNA and protein levels of pro-apoptotic factors Bax and caspase-3 were significantly increased in the miRNA-122 mimics transfection group, while those of anti-apoptotic factor Bcl-2 were significantly decreased compared to those of the control group. The expression of Bax and Caspase-3 decreased, and the expression of anti-apoptotic factor Bcl-2 increased in the transfected miRNA-122 inhibitors group. mRNA expression levels of Hes1, Notch1, VEGF, and CCNG1 in the miRNA-122 mimic transfected group were significantly decreased, while mRNA expression levels in the miRNA-122 inhibitors transfected group were significantly increased. Bioinformatics showed that there was a miRNA-122 binding site in the 3'UTR region of CCNG1, and dual luciferase assay confirmed that CCNG1 was the target of miRNA-122. CONCLUSION Serum miRNA-122 increased significantly after ACI, which may be a diagnostic marker of ACI. miRNA-122 may be involved in the pathological process of ACI and is related to the degree of neurological impairment and short-term prognosis in patients with ACI. miRNA-122 may play a regulatory role in ACI by inhibiting cell proliferation, increasing apoptosis, and inhibiting vascular endothelial cell regeneration through the CCNG1 channel.
Collapse
Affiliation(s)
- Xiao-Juan Yu
- Emergency Department, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, P.R. China
| | - Tian Zhang
- Emergency Department, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, P.R. China
| | - Zeng-Zhen Wei
- Emergency Department, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, P.R. China
| | - Bin Gu
- Emergency Department, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, P.R. China
| | - Ting Guo
- Emergency Department, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, P.R. China
| | - Wen-Juan Jiang
- Emergency Department, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, P.R. China
| | - Yue-Qin Shen
- Emergency Department, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, P.R. China
| | - Dong Wang
- Clinical Laboratory, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, P.R. China
| | - Qian Wang
- Blood Purification Center, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, P.R. China
| | - Jun Wang
- Emergency Department, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, P.R. China.
| |
Collapse
|
77
|
Kittur FS, Hung CY, Li PA, Sane DC, Xie J. Asialo-rhuEPO as a Potential Neuroprotectant for Ischemic Stroke Treatment. Pharmaceuticals (Basel) 2023; 16:610. [PMID: 37111367 PMCID: PMC10143832 DOI: 10.3390/ph16040610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Neuroprotective drugs to protect the brain against cerebral ischemia and reperfusion (I/R) injury are urgently needed. Mammalian cell-produced recombinant human erythropoietin (rhuEPOM) has been demonstrated to have excellent neuroprotective functions in preclinical studies, but its neuroprotective properties could not be consistently translated in clinical trials. The clinical failure of rhuEPOM was thought to be mainly due to its erythropoietic activity-associated side effects. To exploit its tissue-protective property, various EPO derivatives with tissue-protective function only have been developed. Among them, asialo-rhuEPO, lacking terminal sialic acid residues, was shown to be neuroprotective but non-erythropoietic. Asialo-rhuEPO can be prepared by enzymatic removal of sialic acid residues from rhuEPOM (asialo-rhuEPOE) or by expressing human EPO gene in glycoengineered transgenic plants (asialo-rhuEPOP). Both types of asialo-rhuEPO, like rhuEPOM, displayed excellent neuroprotective effects by regulating multiple cellular pathways in cerebral I/R animal models. In this review, we describe the structure and properties of EPO and asialo-rhuEPO, summarize the progress on neuroprotective studies of asialo-rhuEPO and rhuEPOM, discuss potential reasons for the clinical failure of rhuEPOM with acute ischemic stroke patients, and advocate future studies needed to develop asialo-rhuEPO as a multimodal neuroprotectant for ischemic stroke treatment.
Collapse
Affiliation(s)
- Farooqahmed S. Kittur
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| | - Chiu-Yueh Hung
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| | - P. Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| | - David C. Sane
- Carilion Clinic and Virginia Tech Carilion School of Medicine, Roanoke, VA 24014, USA;
| | - Jiahua Xie
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| |
Collapse
|
78
|
Zhou Z, Dun L, Yang Q, Tao J, Yu P, Xu H, Zhao N, Zheng N, An H, Yi P. Tongqiao Huoxue decoction alleviates neurological impairment following ischemic stroke via the PTGS2/NF-kappa B axis. Brain Res 2023; 1805:148247. [PMID: 36669713 DOI: 10.1016/j.brainres.2023.148247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/04/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
Traditional Chinese medicine has emerged as promising targets for ischemic stroke (IS) therapy, yet the mechanism remains elusive. The current study was performed with an aim to investigate the action and mechanism of Tongqiao Huoxue decoction (TQHXD) affecting the neurological impairment secondary to IS based on network pharmacology. Based on network pharmacology and bioinformatics analysis, target genes and pathways involved in the treatment of TQHXD against IS were predicted. Serum containing TQHXD was prepared through blood collection from C57BL/6 mice after intragastric administration of TQHXD. The main results exhibited that Prostaglandin-endoperoxide synthase 2 (PTGS2) exhibited an abundance in IS and enrichment in the NF-kappa B signaling pathway, holding the potential as targets related to TQHXD treatment for IS. TQHXD was found to rescue cell viability, inhibit apoptosis, and alleviate inflammation under oxygen and glucose deprivation and reoxygenation (OGD/R) exposure. Furthermore, our in vivo experiment validated the protective function of TQHXD in ischemic brain damage stimulated by middle cerebral artery occlusion (MCAO). This protective action of TQHXD could be attenuated by overexpressing nuclear factor (NF)-kappa B, which was dependent on PTGS2. Collectively, TQHXD was demonstrated to ameliorate IS-induced neurological impairment by blocking the NF-kappa B signaling pathway and down-regulating PTGS2.
Collapse
Affiliation(s)
- Zheyi Zhou
- Department of Neurology Laboratory, Liuzhou Hospital of Traditional Chinese Medicine, Liuzhou 545001, PR China
| | - Linglu Dun
- Department of Neurology Laboratory, Liuzhou Hospital of Traditional Chinese Medicine, Liuzhou 545001, PR China
| | - Qian Yang
- Department of Neurology Laboratory, Liuzhou Hospital of Traditional Chinese Medicine, Liuzhou 545001, PR China
| | - Jingrui Tao
- Department of Neurology Laboratory, Liuzhou Hospital of Traditional Chinese Medicine, Liuzhou 545001, PR China
| | - Peishan Yu
- Department of Neurology Laboratory, Liuzhou Hospital of Traditional Chinese Medicine, Liuzhou 545001, PR China
| | - Hong Xu
- Department of Neurology Laboratory, Liuzhou Hospital of Traditional Chinese Medicine, Liuzhou 545001, PR China
| | - Na Zhao
- Department of Neurology Laboratory, Liuzhou Hospital of Traditional Chinese Medicine, Liuzhou 545001, PR China
| | - Na Zheng
- Department of Neurology Laboratory, Liuzhou Hospital of Traditional Chinese Medicine, Liuzhou 545001, PR China
| | - Hongwei An
- Department of Neurology Laboratory, Liuzhou Hospital of Traditional Chinese Medicine, Liuzhou 545001, PR China
| | - Ping Yi
- Department of Neurology Laboratory, Liuzhou Hospital of Traditional Chinese Medicine, Liuzhou 545001, PR China.
| |
Collapse
|
79
|
Du Y, Qiu R, Chen L, Chen Y, Zhong Z, Li P, Fan F, Cheng Y. Identification of serum exosomal metabolomic and proteomic profiles for remote ischemic preconditioning. J Transl Med 2023; 21:241. [PMID: 37009888 PMCID: PMC10069038 DOI: 10.1186/s12967-023-04070-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/18/2023] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND Remote ischemic preconditioning (RIPC) refers to a brief episode of exposure to potential adverse stimulation and prevents injury during subsequent exposure. RIPC has been shown to increase tolerance to ischemic injury and improve cerebral perfusion status. Exosomes have a variety of activities, such as remodeling the extracellular matrix and transmitting signals to other cells. This study aimed to investigate the potential molecular mechanism of RIPC-mediated neuroprotection. METHODS Sixty adult male military personnel participants were divided into the control group (n = 30) and the RIPC group (n = 30). We analyzed the differential metabolites and proteins in the serum exosomes of RIPC participants and control subjects. RESULTS Eighty-seven differentially expressed serum exosomal metabolites were found between the RIPC and control groups, which were enriched in pathways related to tyrosine metabolism, sphingolipid metabolism, serotonergic synapses, and multiple neurodegeneration diseases. In addition, there were 75 differentially expressed exosomal proteins between RIPC participants and controls, which involved the regulation of insulin-like growth factor (IGF) transport, neutrophil degranulation, vesicle-mediated transport, etc. Furthermore, we found differentially expressed theobromine, cyclo gly-pro, hemopexin (HPX), and apolipoprotein A1 (ApoA1), which are associated with neuroprotective benefits in ischemia/reperfusion injury. In addition, five potential metabolite biomarkers, including ethyl salicylate, ethionamide, piperic acid, 2, 6-di-tert-butyl-4-hydroxymethylphenol and zerumbone, that separated RIPC from control individuals were identified. CONCLUSION Our data suggest that serum exosomal metabolites are promising biomarkers for RIPC, and our results provide a rich dataset and framework for future analyses of cerebral ischemia‒reperfusion injury under ischemia/reperfusion conditions.
Collapse
Affiliation(s)
- Yang Du
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Rui Qiu
- Institute of National Security, Minzu University of China, Beijing, China
| | - Lei Chen
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Yuewen Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Key Laboratory of Translational Research for Brain Diseases, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhifeng Zhong
- Department of High Altitude Operational Medicine, College of High Altitude Military Medicine, Army Medical University, (Third Military Medical University), Chongqing, China
| | - Peng Li
- Department of High Altitude Operational Medicine, College of High Altitude Military Medicine, Army Medical University, (Third Military Medical University), Chongqing, China
| | - Fangcheng Fan
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China.
| | - Yong Cheng
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China.
- Institute of National Security, Minzu University of China, Beijing, China.
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China.
| |
Collapse
|
80
|
Hayes CA, Morgan NI, Thomas KC, Pushie MJ, Vijayasankar A, Ashmore BG, Wontor K, De Leon MA, Ashpole NM. Neuronal and Astrocyte Insulin-like Growth Factor-1 Signaling Differentially Modulates Ischemic Stroke Damage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.02.535245. [PMID: 37034764 PMCID: PMC10081310 DOI: 10.1101/2023.04.02.535245] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Ischemic stroke is a leading cause of death and disability, as therapeutic options for mitigating the long-term deficits precipitated by the event remain limited. Acute administration of the neuroendocrine modulator insulin-like growth factor-1 (IGF-1) attenuates ischemic stroke damage in preclinical models, and clinical studies suggest IGF-1 can reduce the risk of stroke and improve overall outcomes. The cellular mechanism by which IGF-1 exerts this protection is poorly defined, as all cells within the neurovascular unit express the IGF-1 receptor. We hypothesize that the functional regulation of both neurons and astrocytes by IGF-1 is critical in minimizing damage in ischemic stroke. To test this, we utilized inducible astrocyte-specific or neuron-specific transgenic mouse models to selectively reduce IGF-1R in the adult mouse brain prior to photothrombotic stroke. Acute changes in blood brain barrier permeability, microglial activation, systemic inflammation, and biochemical composition of the brain were assessed 3 hours following photothrombosis, and significant protection was observed in mice deficient in neuronal and astrocytic IGF-1R. When the extent of tissue damage and sensorimotor dysfunction was assessed for 3 days following stroke, only the neurological deficit score continued to show improvements, and the extent of improvement was enhanced with additional IGF-1 supplementation. Overall, results indicate that neuronal and astrocytic IGF-1 signaling influences stroke damage but IGF-1 signaling within these individual cell types is not required for minimizing tissue damage or behavioral outcome.
Collapse
Affiliation(s)
- Cellas A. Hayes
- Department of BioMolecular Sciences, University of Mississippi School of Pharmacy, University of Mississippi, University, MS 386671
| | - Nyah I. Morgan
- Department of BioMolecular Sciences, University of Mississippi School of Pharmacy, University of Mississippi, University, MS 386671
| | - Kamryn C. Thomas
- Department of BioMolecular Sciences, University of Mississippi School of Pharmacy, University of Mississippi, University, MS 386671
| | - M. Jake. Pushie
- Department of Surgery, College of Medicine, University of Saskatchewan, SK S7N 5E5 Canada
| | - Akshaya Vijayasankar
- Department of BioMolecular Sciences, University of Mississippi School of Pharmacy, University of Mississippi, University, MS 386671
| | - Brandon G. Ashmore
- Department of BioMolecular Sciences, University of Mississippi School of Pharmacy, University of Mississippi, University, MS 386671
| | - Kendall Wontor
- Department of Chemistry and Biochemistry, The University of Mississippi, University, Mississippi 38677, United States
| | - Miguel A. De Leon
- Department of BioMolecular Sciences, University of Mississippi School of Pharmacy, University of Mississippi, University, MS 386671
| | - Nicole M. Ashpole
- Department of BioMolecular Sciences, University of Mississippi School of Pharmacy, University of Mississippi, University, MS 386671
- Research Institute of Pharmaceutical Sciences, University of Mississippi School of Pharmacy, University of Mississippi, University, MS 38677
| |
Collapse
|
81
|
Zakeri AS, Wheeler DG, Huttinger A, Carfora A, Kini A, Stork T, Yacoub S, Anderson C, Joseph M, Shujaat MT, Nimjee SM. A canine thromboembolic model of anterior circulation large vessel occlusion stroke. Heliyon 2023; 9:e14692. [PMID: 37089293 PMCID: PMC10119506 DOI: 10.1016/j.heliyon.2023.e14692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/01/2023] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
Purpose To develop a large animal preclinical model of thromboembolic stroke with stable, protracted large vessel occlusion (LVO) utilizing an autologous clot. Materials and methods A reproducible canine model of large vessel occlusion stroke was established by endovascular placement of an autologous clot into the middle cerebral artery (MCA) of six adult hounds and confirmed using digital subtraction angiography (DSA). Infarct volume and evidence of hemorrhage were determined by magnetic resonance imaging (MRI) 7 h after occlusion and Thrombolysis in Cerebral Infarction scale (TICI) was assessed before and after clot placement and at 1, 6, 7, and 9 h after middle cerebral artery occlusion (MCAO). Heart rate (HR) and blood pressure (BP) were monitored continuously and invasively through an arterial sheath throughout the procedures and complete blood count and blood gas analysis completed at time of sacrifice. Histopathological findings at time of sacrifice were used to confirm stroke volume and hemorrhage. Results MCAO with resulting TICI 0 flow was observed in all six animals, verified by serial DSA, and lack of collateral flow persisted for 9 h after clot placement until time of sacrifice. The mean infarct volume was 47.0 ± 6.7% of the ipsilateral hemisphere and no events of spontaneous recanalization or clot autolysis were observed. Conclusion We demonstrate a thromboembolic canine model of MCAO that is both feasible and results in consistent infarct volumes to generate a clinically relevant LVO. This model is important to evaluate treatment of LVO in acute ischemic stroke (AIS) outside the established 4.5 h recombinant tissue plasminogen activator (rTPA) therapeutic window utilizing a prolonged occlusive thrombus.
Collapse
|
82
|
Wei W, Cardes F, Hierlemann A, Modena MM. 3D In Vitro Blood-Brain-Barrier Model for Investigating Barrier Insults. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205752. [PMID: 36782313 PMCID: PMC10104638 DOI: 10.1002/advs.202205752] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/18/2023] [Indexed: 06/18/2023]
Abstract
Blood-brain-barrier (BBB) disruption has been associated with a variety of central-nervous-system diseases. In vitro BBB models enable to investigate how the barrier reacts to external injury events, commonly referred to as insults. Here, a human-cell-based BBB platform with integrated, transparent electrodes to monitor barrier tightness in real time at high resolution is presented. The BBB model includes human cerebral endothelial cells and primary pericytes and astrocytes in a 3D arrangement within a pump-free, open-microfluidic platform. With this platform, this study demonstrates that oxygen-glucose deprivation (OGD), which mimics the characteristics of an ischemic insult, induces a rapid remodeling of the cellular actin structures and subsequent morphological changes in the endothelial cells. High-resolution live imaging shows the formation of large actin stress-fiber bundles in the endothelial layer during OGD application, which ultimately leads to cell shrinkage and barrier breakage. Simultaneous electrical measurements evidence a rapid decrease of the barrier electrical resistance before the appearance of stress fibers, which indicates that the barrier function is compromised already before the appearance of drastic morphological changes. The results demonstrate that the BBB platform recapitulates the main barrier functions in vitro and can be used to investigate rapid reorganization of the BBB upon application of external stimuli.
Collapse
Affiliation(s)
- Wei Wei
- ETH ZürichDepartment of Biosystems Science and EngineeringBio Engineering LaboratoryBasel4058Switzerland
| | - Fernando Cardes
- ETH ZürichDepartment of Biosystems Science and EngineeringBio Engineering LaboratoryBasel4058Switzerland
| | - Andreas Hierlemann
- ETH ZürichDepartment of Biosystems Science and EngineeringBio Engineering LaboratoryBasel4058Switzerland
| | - Mario M. Modena
- ETH ZürichDepartment of Biosystems Science and EngineeringBio Engineering LaboratoryBasel4058Switzerland
| |
Collapse
|
83
|
Huang H, Oo TT, Apaijai N, Chattipakorn N, Chattipakorn SC. An Updated Review of Mitochondrial Transplantation as a Potential Therapeutic Strategy Against Cerebral Ischemia and Cerebral Ischemia/Reperfusion Injury. Mol Neurobiol 2023; 60:1865-1883. [PMID: 36595193 DOI: 10.1007/s12035-022-03200-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023]
Abstract
Regardless of the progress made in the pathogenesis of ischemic stroke, it remains a leading cause of adult disability and death. To date, the most effective treatment for ischemic stroke is the timely recanalization of the occluded artery. However, the short time window and reperfusion injury have greatly limited its application and efficacy. Mitochondrial dysfunction and ATP depletion have become regarded as being hallmarks of neuropathophysiology following ischemic stroke. Mitochondrial transplantation is a novel potential therapeutic intervention for ischemic stroke that has sparked widespread concern during the past few years. This review summarizes and discusses the effects of mitochondrial transplantation in in vitro and in vivo ischemic stroke models. In addition, pharmacological interventions promoting mitochondrial transplantation are reviewed and discussed. We also discuss the potential challenges to the clinical application of mitochondrial transplantation in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Huatuo Huang
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Thura Tun Oo
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nattayaporn Apaijai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand. .,Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand. .,Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
84
|
Oxygen-Glucose Deprivation Decreases the Motility and Length of Axonal Mitochondria in Cultured Dorsal Root Ganglion Cells of Rats. Cell Mol Neurobiol 2023; 43:1267-1280. [PMID: 35771293 DOI: 10.1007/s10571-022-01247-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/20/2022] [Indexed: 11/03/2022]
Abstract
Controlling axonal mitochondria is important for maintaining normal function of the neural network. Oxygen-glucose deprivation (OGD), a model used for mimicking ischemia, eventually induces neuronal cell death similar to axonal degeneration. Axonal mitochondria are disrupted during OGD-induced neural degeneration; however, the mechanism underlying mitochondrial dysfunction has not been completely understood. We focused on the dynamics of mitochondria in axons exposed to OGD; we observed that the number of motile mitochondria significantly reduced in 1 h following OGD exposure. In our observation, the decreased length of stationary mitochondria was affected by the following factors: first, the halt of motile mitochondria; second, the fission of longer stationary mitochondria; and third, a transformation from tubular to spherical shape in OGD-exposed axons. Motile mitochondria reduction preceded stationary mitochondria fragmentation in OGD exposure; these conditions induced the decrease of stationary mitochondria in three different ways. Our results suggest that mitochondrial morphological changes precede the axonal degeneration while ischemia-induced neurodegeneration.
Collapse
|
85
|
Zhang T, Deng D, Huang S, Fu D, Wang T, Xu F, Ma L, Ding Y, Wang K, Wang Y, Zhao W, Chen X. A retrospect and outlook on the neuroprotective effects of anesthetics in the era of endovascular therapy. Front Neurosci 2023; 17:1140275. [PMID: 37056305 PMCID: PMC10086253 DOI: 10.3389/fnins.2023.1140275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Studies on the neuroprotective effects of anesthetics were carried out more than half a century ago. Subsequently, many cell and animal experiments attempted to verify the findings. However, in clinical trials, the neuroprotective effects of anesthetics were not observed. These contradictory results suggest a mismatch between basic research and clinical trials. The Stroke Therapy Academic Industry Roundtable X (STAIR) proposed that the emergence of endovascular thrombectomy (EVT) would provide a proper platform to verify the neuroprotective effects of anesthetics because the haemodynamics of patients undergoing EVT is very close to the ischaemia–reperfusion model in basic research. With the widespread use of EVT, it is necessary for us to re-examine the neuroprotective effects of anesthetics to guide the use of anesthetics during EVT because the choice of anesthesia is still based on team experience without definite guidelines. In this paper, we describe the research status of anesthesia in EVT and summarize the neuroprotective mechanisms of some anesthetics. Then, we focus on the contradictory results between clinical trials and basic research and discuss the causes. Finally, we provide an outlook on the neuroprotective effects of anesthetics in the era of endovascular therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Xiangdong Chen
- *Correspondence: Xiangdong Chen, ; orcid.org/0000-0003-3347-2947
| |
Collapse
|
86
|
Shichita T, Ooboshi H, Yoshimura A. Neuroimmune mechanisms and therapies mediating post-ischaemic brain injury and repair. Nat Rev Neurosci 2023; 24:299-312. [PMID: 36973481 DOI: 10.1038/s41583-023-00690-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2023] [Indexed: 03/29/2023]
Abstract
The nervous and immune systems control whole-body homeostasis and respond to various types of tissue injury, including stroke, in a coordinated manner. Cerebral ischaemia and subsequent neuronal cell death activate resident or infiltrating immune cells, which trigger neuroinflammation that affects functional prognosis after stroke. Inflammatory immune cells exacerbate ischaemic neuronal injury after the onset of brain ischaemia; however, some of the immune cells thereafter change their function to neural repair. The recovery processes after ischaemic brain injury require additional and close interactions between the nervous and immune systems through various mechanisms. Thus, the brain controls its own inflammation and repair processes after injury via the immune system, which provides a promising therapeutic opportunity for stroke recovery.
Collapse
Affiliation(s)
- Takashi Shichita
- Stroke Renaissance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
- Department of Neuroinflammation and Repair, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
- Core Research for Evolutionary Medical Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), Tokyo, Japan.
| | - Hiroaki Ooboshi
- Section of Internal Medicine, Department of Medicine, Fukuoka Dental College, Fukuoka, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
87
|
Kim H, Kim J, Kim J, Oh S, Choi K, Yoon J. Magnetothermal-based non-invasive focused magnetic stimulation for functional recovery in chronic stroke treatment. Sci Rep 2023; 13:4988. [PMID: 36973390 PMCID: PMC10042827 DOI: 10.1038/s41598-023-31979-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Magnetic heat-based brain stimulation of specific lesions could promote the restoration of impaired motor function caused by chronic stroke. We delivered localized stimulation by nanoparticle-mediated heat generation within the targeted brain area via focused magnetic stimulation. The middle cerebral artery occlusion model was prepared, and functional recovery in the chronic-phase stroke rat model was demonstrated by the therapeutic application of focused magnetic stimulation. We observed a transient increase in blood-brain barrier permeability at the target site of < 4 mm and metabolic brain activation at the target lesion. After focused magnetic stimulation, the rotarod score increased by 390 ± 28% (p < 0.05) compared to the control group. Standardized uptake value in the focused magnetic stimulation group increased by 2063 ± 748% (p < 0.01) compared to the control group. Moreover, an increase by 24 ± 5% (p < 0.05) was observed in the sham group as well. Our results show that non-invasive focused magnetic stimulation can safely modulate BBB permeability and enhance neural activation for chronic-phase stroke treatment in the targeted deep brain area.
Collapse
Affiliation(s)
- Hohyeon Kim
- School of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea
| | - Jihye Kim
- Department of Neurology, Chonnam National University Hospital and Medical School, 8 Hak-dong, Dong-gu, Gwangju, 501-757, South Korea
| | - Jahae Kim
- Department of Nuclear Medicines, Chonnam National University Hospital and Medical School, 8 Hak-dong, Dong-gu, Gwangju, 501-757, South Korea
| | - Seungjun Oh
- School of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea
| | - Kangho Choi
- Department of Neurology, Chonnam National University Hospital and Medical School, 8 Hak-dong, Dong-gu, Gwangju, 501-757, South Korea.
| | - Jungwon Yoon
- School of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea.
| |
Collapse
|
88
|
Sunil S, Jiang J, Shah S, Kura S, Kilic K, Erdener SE, Ayata C, Devor A, Boas DA. Neurovascular coupling is preserved in chronic stroke recovery after targeted photothrombosis. Neuroimage Clin 2023; 38:103377. [PMID: 36948140 PMCID: PMC10034641 DOI: 10.1016/j.nicl.2023.103377] [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: 10/13/2022] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
Functional neuroimaging, which measures hemodynamic responses to brain activity, has great potential for monitoring recovery in stroke patients and guiding rehabilitation during recovery. However, hemodynamic responses after stroke are almost always altered relative to responses in healthy subjects and it is still unclear if these alterations reflect the underlying brain physiology or if the alterations are purely due to vascular injury. In other words, we do not know the effect of stroke on neurovascular coupling and are therefore limited in our ability to use functional neuroimaging to accurately interpret stroke pathophysiology. To address this challenge, we simultaneously captured neural activity, through fluorescence calcium imaging, and hemodynamics, through intrinsic optical signal imaging, during longitudinal stroke recovery. Our data suggest that neurovascular coupling was preserved in the chronic phase of recovery (2 weeks and 4 weeks post-stoke) and resembled pre-stroke neurovascular coupling. This indicates that functional neuroimaging faithfully represents the underlying neural activity in chronic stroke. Further, neurovascular coupling in the sub-acute phase of stroke recovery was predictive of long-term behavioral outcomes. Stroke also resulted in increases in global brain oscillations, which showed distinct patterns between neural activity and hemodynamics. Increased neural excitability in the contralesional hemisphere was associated with increased contralesional intrahemispheric connectivity. Additionally, sub-acute increases in hemodynamic oscillations were associated with improved sensorimotor outcomes. Collectively, these results support the use of hemodynamic measures of brain activity post-stroke for predicting functional and behavioral outcomes.
Collapse
Affiliation(s)
- Smrithi Sunil
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| | - John Jiang
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Shashwat Shah
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Sreekanth Kura
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Kivilcim Kilic
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Sefik Evren Erdener
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Cenk Ayata
- Departments of Neurology and Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Anna Devor
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA 02114, USA
| | - David A Boas
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
89
|
Schädlich IS, Winzer R, Stabernack J, Tolosa E, Magnus T, Rissiek B. The role of the ATP-adenosine axis in ischemic stroke. Semin Immunopathol 2023:10.1007/s00281-023-00987-3. [PMID: 36917241 DOI: 10.1007/s00281-023-00987-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/25/2023] [Indexed: 03/16/2023]
Abstract
In ischemic stroke, the primary neuronal injury caused by the disruption of energy supply is further exacerbated by secondary sterile inflammation. The inflammatory cascade is largely initiated by the purine adenosine triphosphate (ATP) which is extensively released to the interstitial space during brain ischemia and functions as an extracellular danger signaling molecule. By engaging P2 receptors, extracellular ATP activates microglia leading to cytokine and chemokine production and subsequent immune cell recruitment from the periphery which further amplifies post-stroke inflammation. The ectonucleotidases CD39 and CD73 shape and balance the inflammatory environment by stepwise degrading extracellular ATP to adenosine which itself has neuroprotective and anti-inflammatory signaling properties. The neuroprotective effects of adenosine are mainly mediated through A1 receptors and inhibition of glutamatergic excitotoxicity, while the anti-inflammatory capacities of adenosine have been primarily attributed to A2A receptor activation on infiltrating immune cells in the subacute phase after stroke. In this review, we summarize the current state of knowledge on the ATP-adenosine axis in ischemic stroke, discuss contradictory results, and point out potential pitfalls towards translating therapeutic approaches from rodent stroke models to human patients.
Collapse
Affiliation(s)
- Ines Sophie Schädlich
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Riekje Winzer
- Institute of Immunology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Joschi Stabernack
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Eva Tolosa
- Institute of Immunology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Björn Rissiek
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| |
Collapse
|
90
|
Wang SN, Wang Z, Wang XY, Zhang XP, Xu TY, Miao CY. Humanized cerebral organoids-based ischemic stroke model for discovering of potential anti-stroke agents. Acta Pharmacol Sin 2023; 44:513-523. [PMID: 36100766 PMCID: PMC9958103 DOI: 10.1038/s41401-022-00986-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/19/2022] [Indexed: 11/09/2022] Open
Abstract
Establishing a stoke experimental model, which is better in line with the physiology and function of human brain, is the bottleneck for the development of effective anti-stroke drugs. A three-dimensional cerebral organoids (COs) from human pluripotent stem cells can mimic cell composition, cortical structure, brain neural connectivity and epigenetic genomics of in-vivo human brain, which provides a promising application in establishing humanized ischemic stroke model. COs have been used for modeling low oxygen condition-induced hypoxic injury, but there is no report on the changes of COs in response to in vitro oxygen-glucose deprivation (OGD)-induced damage of ischemic stroke as well as its application in testing anti-stroke drugs. In this study we compared the cell composition of COs at different culture time and explored the cell types, cell ratios and volume size of COs at 85 days (85 d-CO). The 85 d-CO with diameter more than 2 mm was chosen for establishing humanized ischemic stroke model of OGD. By determining the time-injury relationship of the model, we observed aggravated ischemic injury of COs with OGD exposure time, obtaining first-hand evidence for the damage degree of COs under different OGD condition. The sensitivity of the model to ischemic injury and related treatment was validated by the proven pan-Caspase inhibitor Z-VAD-FMK (20 μM) and Bcl-2 inhibitor navitoclax (0.5 μM). Neuroprotective agents edaravone, butylphthalide, P7C3-A20 and ZL006 (10 μM for each) exerted similar beneficial effects in this model. Taken together, this study establishes a humanized ischemic stroke model based on COs, and provides evidence as a new research platform for anti-stroke drug development.
Collapse
Affiliation(s)
- Shu-Na Wang
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Zhi Wang
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Xi-Yuan Wang
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Xiu-Ping Zhang
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Tian-Ying Xu
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China.
- Department of Anesthesia Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China.
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
91
|
Tong S, Zhong J, Chen X, Deng X, Huang J, Zhang Y, Qin M, Li Z, Cheng H, Zhang W, Zheng L, Xie W, Qiu P, Wang K. In Vivo Deep-Brain 3- and 4-Photon Fluorescence Imaging of Subcortical Structures Labeled by Quantum Dots Excited at the 2200 nm Window. ACS NANO 2023; 17:3686-3695. [PMID: 36799427 DOI: 10.1021/acsnano.2c10724] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Multiphoton microscopy (MPM) is an enabling technology for visualizing deep-brain structures at high spatial resolution in vivo. Within the low tissue absorption window, shifting to longer excitation wavelengths reduces tissue scattering and boosts penetration depth. Recently, the 2200 nm excitation window has emerged as the last and longest window suitable for deep-brain MPM. However, multiphoton fluorescence imaging at this window has not been demonstrated, due to the lack of characterization of multiphoton properties of fluorescent labels. Here we demonstrate technologies for measuring both the multiphoton excitation and emission properties of fluorescent labels at the 2200 nm window, using (1) 3-photon (ησ3) and 4-photon action cross sections (ησ4) and (2) 3-photon and 4-photon emission spectra both ex vivo and in vivo of quantum dots. Our results show that quantum dots have exceptionally large ησ3 and ησ4 for efficient generation of multiphoton fluorescence. Besides, the 3-photon and 4-photon emission spectra of quantum dots are essentially identical to those of one-photon emission, which change negligibly subject to the local environment of circulating blood. Based on these characterization results, we further demonstrate deep-brain vasculature imaging in vivo. Due to the superb multiphoton properties of quantum dots, 3-photon and 4-photon fluorescence imaging reaches a maximum brain imaging depth of 1060 and 940 μm below the surface of a mouse brain, respectively, which enables the imaging of subcortical structures. We thus fill the last gap in multiphoton fluorescence imaging in terms of wavelength selection.
Collapse
Affiliation(s)
- Shen Tong
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jincheng Zhong
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xinlin Chen
- Advanced Life Imaging Lab, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Xiangquan Deng
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jie Huang
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Yingxian Zhang
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Mengyuan Qin
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Zhenhui Li
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Hui Cheng
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Wanjian Zhang
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Lei Zheng
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Weixin Xie
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Ping Qiu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Ke Wang
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
92
|
Hou JY, Zhou XL, Wang XY, Liang J, Xue Q. Peroxiredoxin-6 Released by Astrocytes Contributes to Neuroapoptosis During Ischemia. Neuroscience 2023; 512:59-69. [PMID: 36642396 DOI: 10.1016/j.neuroscience.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 12/26/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023]
Abstract
Peroxiredoxin-6 (PRDX6), a member of the peroxiredoxin family, has progressively emerged as a possible therapeutic target for a variety of brain diseases, particularly Alzheimer's disease and ischemic stroke. However, the role of PRDX6 in neurons under ischemic conditions has remained elusive. Here, we found that astrocytes could release PRDX6 extracellularly after OGD/R, and that PRDX6 release actually worsened neuroapoptosis under OGD/R. We discovered a unique PRDX6/RAGE/JNK signaling pathway that contributes to the effect of neuroapoptosis. We applied a specific inhibitor of the RAGE signaling pathway in a mouse MCAO model and observed significant alterations in animal behavior. Considered together, our findings show the crucial role of the astrocyte-released PRDX6 in the process of neuroapoptosis caused by OGD/R, and could provide novel insights for investigating the molecular mechanism of protecting brain function from ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Jin-Yi Hou
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Department of Acupuncture and Moxibustion, Xuzhou Hospital of Traditional Chinese Medicine, Xuzhou, Jiangsu 221002, China
| | - Xiao-Ling Zhou
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Xiao-Yuan Wang
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Jia Liang
- Department of Neurobiology, Xuzhou Medical University, Xuzhou 221002, China
| | - Qun Xue
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China.
| |
Collapse
|
93
|
Mavroudakis L, Lanekoff I. Ischemic Stroke Causes Disruptions in the Carnitine Shuttle System. Metabolites 2023; 13:metabo13020278. [PMID: 36837897 PMCID: PMC9968086 DOI: 10.3390/metabo13020278] [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: 12/22/2022] [Revised: 01/20/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023] Open
Abstract
Gaining a deep understanding of the molecular mechanisms underlying ischemic stroke is necessary to develop treatment alternatives. Ischemic stroke is known to cause a cellular energy imbalance when glucose supply is deprived, enhancing the role for energy production via β-oxidation where acylcarnitines are essential for the transportation of fatty acids into the mitochondria. Although traditional bulk analysis methods enable sensitive detection of acylcarnitines, they do not provide information on their abundances in various tissue regions. However, with quantitative mass spectrometry imaging the detected concentrations and spatial distributions of endogenous molecules can be readily obtained in an unbiased way. Here, we use pneumatically assisted nanospray desorption electrospray ionization mass spectrometry imaging (PA nano-DESI MSI) doped with internal standards to study the distributions of acylcarnitines in mouse brain affected by stroke. The internal standards enable quantitative imaging and annotation of endogenous acylcarnitines is achieved by studying fragmentation patterns. We report a significant accumulation of long-chain acylcarnitines due to ischemia in brain tissue of the middle cerebral artery occlusion (MCAO) stroke model. Further, we estimate activities of carnitine transporting enzymes and demonstrate disruptions in the carnitine shuttle system that affects the β-oxidation in the mitochondria. Our results show the importance for quantitative monitoring of metabolite distributions in distinct tissue regions to understand cell compensation mechanisms involved in handling damage caused by stroke.
Collapse
|
94
|
Kosugi A, Saga Y, Kudo M, Koizumi M, Umeda T, Seki K. Time course of recovery of different motor functions following a reproducible cortical infarction in non-human primates. Front Neurol 2023; 14:1094774. [PMID: 36846141 PMCID: PMC9947718 DOI: 10.3389/fneur.2023.1094774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/12/2023] [Indexed: 02/11/2023] Open
Abstract
A major challenge in human stroke research is interpatient variability in the extent of sensorimotor deficits and determining the time course of recovery following stroke. Although the relationship between the extent of the lesion and the degree of sensorimotor deficits is well established, the factors determining the speed of recovery remain uncertain. To test these experimentally, we created a cortical lesion over the motor cortex using a reproducible approach in four common marmosets, and characterized the time course of recovery by systematically applying several behavioral tests before and up to 8 weeks after creation of the lesion. Evaluation of in-cage behavior and reach-to-grasp movement revealed consistent motor impairments across the animals. In particular, performance in reaching and grasping movements continued to deteriorate until 4 weeks after creation of the lesion. We also found consistent time courses of recovery across animals for in-cage and grasping movements. For example, in all animals, the score for in-cage behaviors showed full recovery at 3 weeks after creation of the lesion, and the performance of grasping movement partially recovered from 4 to 8 weeks. In addition, we observed longer time courses of recovery for reaching movement, which may rely more on cortically initiated control in this species. These results suggest that different recovery speeds for each movement could be influenced by what extent the cortical control is required to properly execute each movement.
Collapse
Affiliation(s)
- Akito Kosugi
- Department of Neurophysiology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yosuke Saga
- Department of Neurophysiology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Moeko Kudo
- Department of Neurophysiology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Masashi Koizumi
- Department of Neurophysiology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Tatsuya Umeda
- Department of Neurophysiology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan,Department of Integrated Neuroanatomy and Neuroimaging, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuhiko Seki
- Department of Neurophysiology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan,*Correspondence: Kazuhiko Seki ✉
| |
Collapse
|
95
|
Britsch DRS, Syeara N, Stowe AM, Karamyan VT. Rodent Stroke Models to Study Functional Recovery and Neural Repair. Methods Mol Biol 2023; 2616:3-12. [PMID: 36715922 DOI: 10.1007/978-1-0716-2926-0_1] [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] [Indexed: 01/31/2023]
Abstract
Rodent ischemic stroke models are essential research tools for studying this highly prevalent disease and represent a critical element in the translational pipeline for development of new therapies. The majority of ischemic stroke models have been developed to study the acute phase of the disease and neuroprotective strategies, but a subset of models is better suited for studying stroke recovery. Each model therefore has characteristics that lend itself to certain types of investigations and outcome measures, and it is important to consider both explicit and implicit details when designing experiments that utilize each model. The following chapter briefly summarizes the known aspects of the main rodent stroke models with emphasis on their clinical relevance and suitability for studying recovery and neural repair following stroke.
Collapse
Affiliation(s)
- Daimen R S Britsch
- Department of Neurology, Department of Neuroscience, The University of Kentucky, Lexington, KY, USA
| | - Nausheen Syeara
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Ann M Stowe
- Department of Neurology, Department of Neuroscience, The University of Kentucky, Lexington, KY, USA
| | - Vardan T Karamyan
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA.
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI, USA.
| |
Collapse
|
96
|
Kim MK, Choi W, Moon HJ, Han S, Shin HJ. Targeted photothrombotic subcortical small vessel occlusion using in vivo real-time fiber bundle endomicroscopy in mice. BIOMEDICAL OPTICS EXPRESS 2023; 14:687-702. [PMID: 36874485 PMCID: PMC9979683 DOI: 10.1364/boe.473407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/18/2022] [Accepted: 12/23/2022] [Indexed: 06/18/2023]
Abstract
The development of an accurate subcortical small vessel occlusion model for pathophysiological studies of subcortical ischemic stroke is still insignificant. In this study, in vivo real-time fiber bundle endomicroscopy (FBEµ) was applied to develop subcortical photothrombotic small vessel occlusion model in mice with minimal invasiveness. Our FBFµ system made it possible to precisely target specific blood vessels in deep brain and simultaneously observe the clot formation and blood flow blockage inside the target blood vessel during photochemical reactions. A fiber bundle probe was directly inserted into the anterior pretectal nucleus of the thalamus in brain of live mice to induce a targeted occlusion in small vessels. Then, targeted photothrombosis was performed using a patterned laser, observing the process through the dual-color fluorescence imaging. On day one post occlusion, infarct lesions are measured using TTC staining and post hoc histology. The results show that FBEµ applied to targeted photothrombosis can successfully generate a subcortical small vessel occlusion model for lacunar stroke.
Collapse
Affiliation(s)
- Min-kyung Kim
- Bionics Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Wonseok Choi
- Bionics Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of Biomedical Engineering, Yonsei University, 1 Yonseidae-gil, Wonju, Gangwon-do, 26493, Republic of Korea
| | - Hyuk-June Moon
- Bionics Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Sungmin Han
- Bionics Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hyun-joon Shin
- Bionics Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| |
Collapse
|
97
|
Shibuya N, Itokazu T, Ueda T, Yamashita T. Intravital Imaging Reveals the Ameliorating Effect of Colchicine in a Photothrombotic Stroke Model via Inhibition of Neutrophil Recruitment. Transl Stroke Res 2023; 14:100-110. [PMID: 35441983 DOI: 10.1007/s12975-022-01022-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 04/12/2022] [Accepted: 04/12/2022] [Indexed: 01/31/2023]
Abstract
Although post-stroke neutrophil recruitment is known to be deleterious to neural tissues in the peri-infarct area, the precise behavior of recruited neutrophils remains elusive. In this study, potential therapeutic agents for modifying neutrophil behavior in the peri-infarct area were explored through intravital imaging of an experimental stroke mouse model. By applying in vivo 2-photon imaging to study a tightly controlled photothrombotic stroke mouse model, we established a highly sensitive and reproducible method for investigating the temporal dynamics of ischemic brain lesions. Taking advantage of this system, we revealed that neutrophil depletion by a neutrophil-specific antibody ameliorated the expansion of the infarct area, confirming the deleterious effect of neutrophils in the peri-infarct cortex. To identify neutrophil-targeted therapeutic approaches, we screened various agents and found that colchicine and an anti-P-selectin antibody were the most effective in inhibiting neutrophil attachment to the vessel wall in the early phase (6 h post-infarction). Interestingly, further investigation in the later phase (16 h post-infarction) revealed that colchicine potently inhibited neutrophil infiltration into the peri-infarct cortex; however, the anti-P-selectin antibody did not. Subsequent analysis revealed that the effect of the anti-P-selectin antibody against neutrophil attachment to the vessel wall was transient and thus insufficient for mitigating neutrophil infiltration. Finally, we revealed that colchicine treatment effectively ameliorated infarct expansion. In conclusion, we have established an intravital strategy to directly investigate pathophysiology in the ischemic border zone, and found that colchicine administration in the acute phase of ischemic stroke is a potential novel therapeutic strategy.
Collapse
Affiliation(s)
- Nao Shibuya
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Takahide Itokazu
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan.
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.
| | - Tsubasa Ueda
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan.
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.
- WPI-Immunology Frontier Research Center, Osaka University, Suita, Japan.
| |
Collapse
|
98
|
Xu S, Li X, Wang Y. Regulation of the p53‑mediated ferroptosis signaling pathway in cerebral ischemia stroke (Review). Exp Ther Med 2023; 25:113. [PMID: 36793330 PMCID: PMC9922943 DOI: 10.3892/etm.2023.11812] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/16/2022] [Indexed: 01/27/2023] Open
Abstract
Stroke is one of the most threatening diseases worldwide, particularly in countries with larger populations; it is associated with high morbidity, mortality and disability rates. As a result, extensive research efforts are being made to address these issues. Stroke can include either hemorrhagic stroke (blood vessel ruptures) or ischemic stroke (blockage of an artery). Whilst the incidence of stroke is higher in the elderly population (≥65), it is also increasing in the younger population. Ischemic stroke accounts for ~85% of all stroke cases. The pathogenesis of cerebral ischemic injury can include inflammation, excitotoxic injury, mitochondrial dysfunction, oxidative stress, ion imbalance and increased vascular permeability. All of the aforementioned processes have been extensively studied, providing insights into the disease. Other clinical consequences observed include brain edema, nerve injury, inflammation, motor deficits and cognitive impairment, which not only cause disabilities obstructing daily life but also increase the mortality rates. Ferroptosis is a type of cell death that is characterized by iron accumulation and increased lipid peroxidation in cells. In particular, ferroptosis has been previously implicated in ischemia-reperfusion injury in the central nervous system. It has also been identified as a mechanism involved in cerebral ischemic injury. The tumor suppressor p53 has been reported to modulate the ferroptotic signaling pathway, which both positively and negatively affects the prognosis of cerebral ischemia injury. The present review summarizes the recent findings on the molecular mechanisms of ferroptosis under the regulation of p53 underlying cerebral ischemia injury. Understanding of the p53/ferroptosis signaling pathway may provide insights into developing methods for improving the diagnosis, treatment and even prevention of stroke.
Collapse
Affiliation(s)
- Shuangli Xu
- Department of Emergency, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Xuewei Li
- Department of Rheumatology and Immunology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Yanqiang Wang
- Department of Neurology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China,Correspondence to: Dr Yanqiang Wang, Department of Neurology, Affiliated Hospital of Weifang Medical University, 2,428 Yuhe Road, Kuiwen, Weifang, Shandong 261031, P.R. China
| |
Collapse
|
99
|
Wang YY, Lin SY, Chang CY, Wu CC, Chen WY, Huang WC, Liao SL, Wang WY, Chen CJ. α7 nicotinic acetylcholine receptor agonist improved brain injury and impaired glucose metabolism in a rat model of ischemic stroke. Metab Brain Dis 2023; 38:1249-1259. [PMID: 36662413 DOI: 10.1007/s11011-023-01167-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 01/13/2023] [Indexed: 01/21/2023]
Abstract
Vagus nerve stimulation through the action of acetylcholine can modulate inflammatory responses and metabolism. α7 Nicotinic Acetylcholine Receptor (α7nAChR) is a key component in the biological functions of acetylcholine. To further explore the health benefits of vagus nerve stimulation, this study aimed to investigate whether α7nAChR agonists offer beneficial effects against poststroke inflammatory and metabolic changes and to identify the underlying mechanisms in a rat model of stroke established by permanent cerebral ischemia. We found evidence showing that pretreatment with α7nAChR agonist, GTS-21, improved poststroke brain infarction size, impaired motor coordination, brain apoptotic caspase 3 activation, dysregulated glucose metabolism, and glutathione reduction. In ischemic cortical tissues and gastrocnemius muscles with GTS-21 pretreatment, macrophages/microglia M1 polarization-associated Tumor Necrosis Factor-α (TNF-α) mRNA, Cluster of Differentiation 68 (CD68) protein, and Inducible Nitric Oxide Synthase (iNOS) protein expression were reduced, while expression of anti-inflammatory cytokine IL-4 mRNA, and levels of M2 polarization-associated CD163 mRNA and protein were increased. In the gastrocnemius muscles, stroke rats showed a reduction in both glutathione content and Akt Serine 473 phosphorylation, as well as an elevation in Insulin Receptor Substrate-1 Serine 307 phosphorylation and Dynamin-Related Protein 1 Serine 616 phosphorylation. GTS-21 reversed poststroke changes in the gastrocnemius muscles. Overall, our findings, provide further evidence supporting the neuroprotective benefits of α7nAChR agonists, and indicate that they may potentially exert anti-inflammatory and metabolic effects peripherally in the skeletal muscle in an acute ischemic stroke animal model.
Collapse
Affiliation(s)
- Ya-Yu Wang
- Department of Family Medicine, Taichung Veterans General Hospital, 407, Taichung City, Taiwan
| | - Shih-Yi Lin
- Center for Geriatrics and Gerontology, Taichung Veterans General Hospital, 407, Taichung City, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, 112, Taipei City, Taiwan
| | - Cheng-Yi Chang
- Department of Surgery, Feng Yuan Hospital, 420, Taichung City, Taiwan
- Department of Veterinary Medicine, National Chung Hsing University, 402, Taichung City, Taiwan
| | - Chih-Cheng Wu
- Department of Anesthesiology, Taichung Veterans General Hospital, 407, Taichung City, Taiwan
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, 402, Taichung City, Taiwan
| | - Wei-Chi Huang
- Department of Veterinary Medicine, National Chung Hsing University, 402, Taichung City, Taiwan
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, No. 1650, Sec. 4, Taiwan Boulevard, 407, Taichung City, Taiwan
| | - Wen-Yi Wang
- Department of Nursing, Hung Kuang University, 433, Taichung City, Taiwan
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, No. 1650, Sec. 4, Taiwan Boulevard, 407, Taichung City, Taiwan.
- Department of Medical Laboratory Science and Biotechnology, China Medical University, 404, Taichung City, Taiwan.
| |
Collapse
|
100
|
Löscher W, Stafstrom CE. Epilepsy and its neurobehavioral comorbidities: Insights gained from animal models. Epilepsia 2023; 64:54-91. [PMID: 36197310 DOI: 10.1111/epi.17433] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/04/2022] [Accepted: 10/04/2022] [Indexed: 01/21/2023]
Abstract
It is well established that epilepsy is associated with numerous neurobehavioral comorbidities, with a bidirectional relationship; people with epilepsy have an increased incidence of depression, anxiety, learning and memory difficulties, and numerous other psychosocial challenges, and the occurrence of epilepsy is higher in individuals with those comorbidities. Although the cause-and-effect relationship is uncertain, a fuller understanding of the mechanisms of comorbidities within the epilepsies could lead to improved therapeutics. Here, we review recent data on epilepsy and its neurobehavioral comorbidities, discussing mainly rodent models, which have been studied most extensively, and emphasize that clinically relevant information can be gained from preclinical models. Furthermore, we explore the numerous potential factors that may confound the interpretation of emerging data from animal models, such as the specific seizure induction method (e.g., chemical, electrical, traumatic, genetic), the role of species and strain, environmental factors (e.g., laboratory environment, handling, epigenetics), and the behavioral assays that are chosen to evaluate the various aspects of neural behavior and cognition. Overall, the interplay between epilepsy and its neurobehavioral comorbidities is undoubtedly multifactorial, involving brain structural changes, network-level differences, molecular signaling abnormalities, and other factors. Animal models are well poised to help dissect the shared pathophysiological mechanisms, neurological sequelae, and biomarkers of epilepsy and its comorbidities.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Carl E Stafstrom
- Division of Pediatric Neurology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|