1
|
McDonough A, Weinstein JR. Glial 'omics in ischemia: Acute stroke and chronic cerebral small vessel disease. Glia 2024. [PMID: 39463002 DOI: 10.1002/glia.24634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/17/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024]
Abstract
Vascular injury and pathologies underlie common diseases including ischemic stroke and cerebral small vessel disease (CSVD). Prior work has identified a key role for glial cells, including microglia, in the multifaceted and temporally evolving neuroimmune response to both stroke and CSVD. Transcriptional profiling has led to important advances including identification of distinct gene expression signatures in ischemia-exposed, flow cytometrically sorted microglia and more recently single cell RNA sequencing-identified microglial subpopulations or clusters. There is a reassuring degree of overlap in the results from these two distinct methodologies with both identifying a proliferative and a separate type I interferon responsive microglial element. Similar patterns were later seen using multimodal and spatial transcriptomal profiling in ischemia-exposed microglia and astrocytes. Methodological advances including enrichment of specific neuroanatomic/functional regions (such as the neurovascular unit) prior to single cell RNA sequencing has led to identification of novel cellular subtypes and generation of new credible hypotheses as to cellular function based on the enhanced cell sub-type specific gene expression patterns. A ribosomal tagging strategy focusing on the cellular translatome analyses carried out in the acute phases post stroke has revealed distinct inflammation-regulating roles for microglia and astrocytes in this setting. Early spatial transcriptomics experiments using cerebral ischemia models have identified regionally distinct microglial cell clusters in ischemic core versus penumbra. There is great potential for combination of these methods for multi-omics approaches to further elucidate glial responses in the context of both acute ischemic stroke and chronic CSVD.
Collapse
Affiliation(s)
- Ashley McDonough
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington, USA
| | - Jonathan R Weinstein
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington, USA
- Department of Neurological Surgery, School of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
2
|
Zhao J, Liu S, Li K, Yang Y, Zhao Y, Zhu X. RBM3 Promotes Anti-inflammatory Responses in Microglia and Serves as a Neuroprotective Target of Ischemic Stroke. Mol Neurobiol 2024; 61:7384-7402. [PMID: 38386136 DOI: 10.1007/s12035-024-04052-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 02/09/2024] [Indexed: 02/23/2024]
Abstract
Ischemic stroke is a major cause of death and disability in adults. Hypothermic treatment is successful in treating neonatal cerebral ischemia, but its application is restricted in adult patients due to complex management strategies and severe adverse effects. Two homologous RNA-binding proteins, RBM3 and CIRP, are the only known cold-inducible proteins in vertebrates, and their expression levels are robustly elevated by mild to moderate hypothermia. In previous studies, we and others have demonstrated that both RBM3 and CIRP mediate the neuroprotective and neurogenic effects of hypothermia in cell and animal models. However, CIRP can also be detrimental to neurons by triggering neuroinflammatory responses, complicating its post-stroke functions. In this study, we compared the properties of the two cold-inducible RNA-binding proteins after ischemic stroke. Our results indicated that RBM3 expression was stimulated in the ischemic brain of stroke patients, while CIRP expression was not. In an experimental model, RBM3 can ameliorate ischemic-like insult by promoting neuronal survival and eliciting anti-inflammatory responses in activated microglia, while the impact of CIRP was intriguing. Collectively, our data supported the notion that RBM3 may be a more promising therapeutic target than CIRP for treating ischemic stroke. We further demonstrated that zr17-2, a small molecule initially identified to target CIRP, can specifically target RBM3 but not CIRP in microglia. zr17-2 demonstrated anti-inflammatory and neuroprotective effects after ischemic stroke both in vitro and in vivo, suggesting its potential therapeutic value.
Collapse
Affiliation(s)
- Junyi Zhao
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen, China
| | - Siyu Liu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen, China
| | - Kunyu Li
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, China
| | - Yulu Yang
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yue Zhao
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen, China
| | - Xinzhou Zhu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, China.
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen, China.
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.
| |
Collapse
|
3
|
Cha LN, Yang J, Gao JA, Lu X, Chang XL, Thuku RC, Liu Q, Lu QM, Li DS, Lai R, Fang MQ. Bat-derived oligopeptide LE6 inhibits the contact-kinin pathway and harbors anti-thromboinflammation and stroke potential. Zool Res 2024; 45:1001-1012. [PMID: 39147715 PMCID: PMC11491786 DOI: 10.24272/j.issn.2095-8137.2023.372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/30/2024] [Indexed: 08/17/2024] Open
Abstract
Thrombosis and inflammation are primary contributors to the onset and progression of ischemic stroke. The contact-kinin pathway, initiated by plasma kallikrein (PK) and activated factor XII (FXIIa), functions bidirectionally with the coagulation and inflammation cascades, providing a novel target for therapeutic drug development in ischemic stroke. In this study, we identified a bat-derived oligopeptide from Myotis myotis (Borkhausen, 1797), designated LE6 (Leu-Ser-Glu-Glu-Pro-Glu, 702 Da), with considerable potential in stroke therapy due to its effects on the contact kinin pathway. Notably, LE6 demonstrated significant inhibitory effects on PK and FXIIa, with inhibition constants of 43.97 μmol/L and 6.37 μmol/L, respectively. In vitro analyses revealed that LE6 prolonged plasma recalcification time and activated partial thromboplastin time. In murine models, LE6 effectively inhibited carrageenan-induced mouse tail thrombosis, FeCl 3-induced carotid artery thrombosis, and photochemically induced intracerebral thrombosis. Furthermore, LE6 significantly decreased inflammation and stroke injury in transient middle cerebral artery occlusion models. Notably, the low toxicity, hemolytic activity, and bleeding risk of LE6, along with its synthetic simplicity, underscore its clinical applicability. In conclusion, as an inhibitor of FXIIa and PK, LE6 offers potential therapeutic benefits in stroke treatment by mitigating inflammation and preventing thrombus formation.
Collapse
Affiliation(s)
- Li-Na Cha
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| | - Juan Yang
- First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Jin-Ai Gao
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| | - Xin Lu
- First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Xiao-Long Chang
- First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Rebecca Caroline Thuku
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Qi Liu
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Qiu-Min Lu
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Dong-Sheng Li
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Ren Lai
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China. E-mail:
| | - Ming-Qian Fang
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China. E-mail:
| |
Collapse
|
4
|
Martins-Silva C, Anderson CL, Boyce AKJ, Andrade TES, Tizziani T, Lopes KHS, Micke GA, Cregan SP, Dos Santos ARS, Thompson RJ. The Ethanolic Extract of Polygala paniculata L. Blocks Panx1 Channels and Reduces Ischemic Brain Infarct in a Dose- and Sex-Dependent Way. Mol Neurobiol 2024:10.1007/s12035-024-04453-5. [PMID: 39271622 DOI: 10.1007/s12035-024-04453-5] [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: 05/27/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
Polygala paniculata L. is a native plant from tropical America. The therapeutic potential of the hydroalcoholic extract of P. paniculata (HEPp) has been scientifically explored due to folk medicine reports on its action against several afflictions. HEPp contains several bioactive molecules with neuroprotective activities, making it a promising candidate for stroke treatment. This study used electrophysiological, biochemical, and in vivo experiments to evaluate the molecular mechanisms underlying HEPp as a neuroprotective therapy for stroke targeting Pannexin-1 (Panx1). Panx1 is a non-selective channel that opens during ischemia and contributes to neuronal death. HEPp was not toxic to cortical neurons and pre-treatment with the extract reduced neuronal death promoted by oxygen and glucose deprivation in a dose-dependent manner. Additionally, HEPp blocked Panx1 currents in a dose-dependent manner and the effect, which was shown to be partially due to rutin. Animals submitted to photothrombosis and post-treated with HEPp had reduced infarct volume, and the effective dose was lower in males (1 mg/kg) than in females (10 mg/kg). On the other hand, in Panx1 KD mice (50% Panx1 levels), the acute treatment reduced the infarct volume only in males. Upon chronic treatment with HEPp, a reduction in Panx1 protein levels was observed. The current study provides reliable evidence of the neuroprotective properties of HEPp in both in vitro and in vivo models of stroke. The underlying mechanism involves, at least in part, the inhibition of Panx1 channel function and possibly downregulation of protein levels, suppressing the secondary events that lead to apoptosis and inflammation.
Collapse
Affiliation(s)
- Cristina Martins-Silva
- Department of Physiological Sciences, Health Sciences Center, Laboratory of Neurochemistry and Behaviour (LabNeC), Graduate Program in Biochemistry, Federal University of Espirito Santo, Vitoria, ES, 29043910, Brazil.
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 1N4, Canada.
| | - Connor L Anderson
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - Andrew K J Boyce
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - Tassiane E S Andrade
- Laboratory of Neurobiology of Pain and Inflammation, Department of Physiological Sciences, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
- Neuroscience Program, Department of Physiology and Pharmacology, University of Western Ontario, Robarts Research Institute, University of Western Ontario, 100 Perth Dr, London, ON, N6A 5K8, Canada
| | - Tiago Tizziani
- Department of Chemistry, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Kheytiany H S Lopes
- Department of Chemistry, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Gustavo A Micke
- Department of Chemistry, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Sean P Cregan
- Neuroscience Program, Department of Physiology and Pharmacology, University of Western Ontario, Robarts Research Institute, University of Western Ontario, 100 Perth Dr, London, ON, N6A 5K8, Canada
| | - Adair Roberto Soares Dos Santos
- Laboratory of Neurobiology of Pain and Inflammation, Department of Physiological Sciences, Biological Sciences Center, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Roger J Thompson
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 1N4, Canada
| |
Collapse
|
5
|
Buchert R, Huppertz HJ, Wegner F, Berding G, Brendel M, Apostolova I, Buhmann C, Poetter-Nerger M, Dierks A, Katzdobler S, Klietz M, Levin J, Mahmoudi N, Rinscheid A, Quattrone A, Rogozinski S, Rumpf JJ, Schneider C, Stoecklein S, Spetsieris PG, Eidelberg D, Sabri O, Barthel H, Wattjes MP, Höglinger G. Added value of FDG-PET for detection of progressive supranuclear palsy. J Neurol Neurosurg Psychiatry 2024:jnnp-2024-333590. [PMID: 39107038 DOI: 10.1136/jnnp-2024-333590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 07/17/2024] [Indexed: 08/09/2024]
Abstract
BACKGROUND Diagnostic criteria for progressive supranuclear palsy (PSP) include midbrain atrophy in MRI and hypometabolism in [18F]fluorodeoxyglucose (FDG)-positron emission tomography (PET) as supportive features. Due to limited data regarding their relative and sequential value, there is no recommendation for an algorithm to combine both modalities to increase diagnostic accuracy. This study evaluated the added value of sequential imaging using state-of-the-art methods to analyse the images regarding PSP features. METHODS The retrospective study included 41 PSP patients, 21 with Richardson's syndrome (PSP-RS), 20 with variant PSP phenotypes (vPSP) and 46 sex- and age-matched healthy controls. A pretrained support vector machine (SVM) for the classification of atrophy profiles from automatic MRI volumetry was used to analyse T1w-MRI (output: MRI-SVM-PSP score). Covariance pattern analysis was applied to compute the expression of a predefined PSP-related pattern in FDG-PET (output: PET-PSPRP expression score). RESULTS The area under the receiver operating characteristic curve for the detection of PSP did not differ between MRI-SVM-PSP and PET-PSPRP expression score (p≥0.63): about 0.90, 0.95 and 0.85 for detection of all PSP, PSP-RS and vPSP. The MRI-SVM-PSP score achieved about 13% higher specificity and about 15% lower sensitivity than the PET-PSPRP expression score. Decision tree models selected the MRI-SVM-PSP score for the first branching and the PET-PSPRP expression score for a second split of the subgroup with normal MRI-SVM-PSP score, both in the whole sample and when restricted to PSP-RS or vPSP. CONCLUSIONS FDG-PET provides added value for PSP-suspected patients with normal/inconclusive T1w-MRI, regardless of PSP phenotype and the methods to analyse the images for PSP-typical features.
Collapse
Affiliation(s)
- Ralph Buchert
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Florian Wegner
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Georg Berding
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Ivayla Apostolova
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Buhmann
- Department of Neurology, University Medical Center Eppendorf, Hamburg, Germany
| | | | - Alexander Dierks
- Department of Nuclear Medicine, University Hospital Augsburg, Augsburg, Germany
| | - Sabrina Katzdobler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Martin Klietz
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Nima Mahmoudi
- Department of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
| | - Andreas Rinscheid
- Medical Physics and Radiation Protection, University Hospital Augsburg, Augsburg, Germany
| | - Andrea Quattrone
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
- Institute of Neurology, Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | | | | | - Christine Schneider
- Department of Neurology and Clinical Neurophysiology, University Hospital Augsburg, Augsburg, Germany
| | - Sophia Stoecklein
- Department of Radiology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Phoebe G Spetsieris
- Feinstein Institutes for Medical Research Manhasset, Manhasset, New York, USA
| | - David Eidelberg
- Feinstein Institutes for Medical Research Manhasset, Manhasset, New York, USA
| | - Osama Sabri
- Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig, Germany
| | - Mike P Wattjes
- Department of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
- Department of Neuroradiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Günter Höglinger
- Department of Neurology, Hannover Medical School, Hannover, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| |
Collapse
|
6
|
Miraglia F, Pappalettera C, Barbati SA, Podda MV, Grassi C, Rossini PM, Vecchio F. Brain complexity in stroke recovery after bihemispheric transcranial direct current stimulation in mice. Brain Commun 2024; 6:fcae137. [PMID: 38741663 PMCID: PMC11089417 DOI: 10.1093/braincomms/fcae137] [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: 09/20/2023] [Revised: 12/22/2023] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
Stroke is one of the leading causes of disability worldwide. There are many different rehabilitation approaches aimed at improving clinical outcomes for stroke survivors. One of the latest therapeutic techniques is the non-invasive brain stimulation. Among non-invasive brain stimulation, transcranial direct current stimulation has shown promising results in enhancing motor and cognitive recovery both in animal models of stroke and stroke survivors. In this framework, one of the most innovative methods is the bihemispheric transcranial direct current stimulation that simultaneously increases excitability in one hemisphere and decreases excitability in the contralateral one. As bihemispheric transcranial direct current stimulation can create a more balanced modulation of brain activity, this approach may be particularly useful in counteracting imbalanced brain activity, such as in stroke. Given these premises, the aim of the current study has been to explore the recovery after stroke in mice that underwent a bihemispheric transcranial direct current stimulation treatment, by recording their electric brain activity with local field potential and by measuring behavioural outcomes of Grip Strength test. An innovative parameter that explores the complexity of signals, namely the Entropy, recently adopted to describe brain activity in physiopathological states, was evaluated to analyse local field potential data. Results showed that stroke mice had higher values of Entropy compared to healthy mice, indicating an increase in brain complexity and signal disorder due to the stroke. Additionally, the bihemispheric transcranial direct current stimulation reduced Entropy in both healthy and stroke mice compared to sham stimulated mice, with a greater effect in stroke mice. Moreover, correlation analysis showed a negative correlation between Entropy and Grip Strength values, indicating that higher Entropy values resulted in lower Grip Strength engagement. Concluding, the current evidence suggests that the Entropy index of brain complexity characterizes stroke pathology and recovery. Together with this, bihemispheric transcranial direct current stimulation can modulate brain rhythms in animal models of stroke, providing potentially new avenues for rehabilitation in humans.
Collapse
Affiliation(s)
- Francesca Miraglia
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele, 00163, Rome, Italy
- Department of Theoretical and Applied Sciences, eCampus University, Novedrate, 22060, Como, Italy
| | - Chiara Pappalettera
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele, 00163, Rome, Italy
- Department of Theoretical and Applied Sciences, eCampus University, Novedrate, 22060, Como, Italy
| | - Saviana Antonella Barbati
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Maria Vittoria Podda
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Paolo Maria Rossini
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele, 00163, Rome, Italy
| | - Fabrizio Vecchio
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele, 00163, Rome, Italy
- Department of Theoretical and Applied Sciences, eCampus University, Novedrate, 22060, Como, Italy
| |
Collapse
|
7
|
Mosberger AC, Sibener LJ, Chen TX, Rodrigues HFM, Hormigo R, Ingram JN, Athalye VR, Tabachnik T, Wolpert DM, Murray JM, Costa RM. Exploration biases forelimb reaching strategies. Cell Rep 2024; 43:113958. [PMID: 38520691 PMCID: PMC11097405 DOI: 10.1016/j.celrep.2024.113958] [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: 07/10/2023] [Revised: 12/05/2023] [Accepted: 02/28/2024] [Indexed: 03/25/2024] Open
Abstract
The brain can generate actions, such as reaching to a target, using different movement strategies. We investigate how such strategies are learned in a task where perched head-fixed mice learn to reach to an invisible target area from a set start position using a joystick. This can be achieved by learning to move in a specific direction or to a specific endpoint location. As mice learn to reach the target, they refine their variable joystick trajectories into controlled reaches, which depend on the sensorimotor cortex. We show that individual mice learned strategies biased to either direction- or endpoint-based movements. This endpoint/direction bias correlates with spatial directional variability with which the workspace was explored during training. Model-free reinforcement learning agents can generate both strategies with similar correlation between variability during training and learning bias. These results provide evidence that reinforcement of individual exploratory behavior during training biases the reaching strategies that mice learn.
Collapse
Affiliation(s)
- Alice C Mosberger
- Departments of Neuroscience and Neurology, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA.
| | - Leslie J Sibener
- Departments of Neuroscience and Neurology, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Tiffany X Chen
- Departments of Neuroscience and Neurology, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Helio F M Rodrigues
- Departments of Neuroscience and Neurology, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA; Allen Institute, Seattle, WA 98109, USA
| | - Richard Hormigo
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - James N Ingram
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Vivek R Athalye
- Departments of Neuroscience and Neurology, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Tanya Tabachnik
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Daniel M Wolpert
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - James M Murray
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | - Rui M Costa
- Departments of Neuroscience and Neurology, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA; Allen Institute, Seattle, WA 98109, USA.
| |
Collapse
|
8
|
Mbs GBY, Wasek B, Bottiglieri T, Malysheva O, Caudill MA, Jadavji NM. Dietary vitamin B12 deficiency impairs motor function and changes neuronal survival and choline metabolism after ischemic stroke in middle-aged male and female mice. Nutr Neurosci 2024; 27:300-309. [PMID: 36932327 DOI: 10.1080/1028415x.2023.2188639] [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: 03/19/2023]
Abstract
Nutrition is a modifiable risk factor for ischemic stroke. As people age their ability to absorb some nutrients decreases, a primary example is vitamin B12. Older individuals with a vitamin B12 deficiency are at a higher risk for ischemic stroke and have worse stroke outcome. However, the mechanisms through which these occur remain unknown. The aim of the study was to investigate the role of vitamin B12 deficiency in ischemic stroke outcome and mechanistic changes in a mouse model. Ten-month-old male and female mice were put on control or vitamin B12 deficient diets for 4 weeks prior to and after ischemic stroke to the sensorimotor cortex. Motor function was measured, and tissues were collected to assess potential mechanisms. All deficient mice had increased levels of total homocysteine in plasma and liver tissues. After ischemic stroke, deficient mice had impaired motor function compared to control mice. There was no difference between groups in ischemic damage volume. However, within the ischemic damage region, there was an increase in total apoptosis of male deficient mice compared to controls. Furthermore, there was an increase in neuronal survival in ischemic brain tissue of the vitamin B12 deficient mice compared to controls. Additionally, there were changes in choline metabolites in ischemic brain tissue because of a vitamin B12 deficiency. The data presented in this study confirms that a vitamin B12 deficiency worsens stroke outcome in male and female mice. The mechanisms driving this change may be a result of neuronal survival and compensation in choline metabolism within the damaged brain tissue.
Collapse
Affiliation(s)
- Gyllian B Yahn Mbs
- Department of Biomedical Sciences, Midwestern University, Glendale, AZ, USA
| | - Brandi Wasek
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott & White Research Institute, Dallas, TX, USA
| | - Teodoro Bottiglieri
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott & White Research Institute, Dallas, TX, USA
| | - Olga Malysheva
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Marie A Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Nafisa M Jadavji
- Department of Biomedical Sciences, Midwestern University, Glendale, AZ, USA
- College of Veterinary Medicine, Midwestern University, Glendale, AZ, USA
- College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
- Department of Neuroscience, Carleton University, Ottawa, Canada
| |
Collapse
|
9
|
Liu L, Ding M, Wu J, Zhang Y, Wang Q, Wang N, Luo L, Yu K, Fan Y, Zhang J, Wu Y, Xiao X, Zhang Q. High-frequency repetitive transcranial magnetic stimulation promotes ipsilesional functional hyperemia and motor recovery in mice with ischemic stroke. Cereb Cortex 2024; 34:bhae074. [PMID: 38511722 DOI: 10.1093/cercor/bhae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 03/22/2024] Open
Abstract
Neurovascular decoupling plays a significant role in dysfunction following an ischemic stroke. This study aimed to explore the effect of low- and high-frequency repetitive transcranial magnetic stimulation on neurovascular remodeling after ischemic stroke. To achieve this goal, we compared functional hyperemia, cerebral blood flow regulatory factors, and neurochemical transmitters in the peri-infract cortex 21 days after a photothrombotic stroke. Our findings revealed that low- and high-frequency repetitive transcranial magnetic stimulation increased the real-time cerebral blood flow in healthy mice and improved neurobehavioral outcomes after stroke. Furthermore, high-frequency (5-Hz) repetitive transcranial magnetic stimulation revealed stronger functional hyperemia recovery and increased the levels of post-synaptic density 95, neuronal nitric oxide synthase, phosphorylated-endothelial nitric oxide synthase, and vascular endothelial growth factor in the peri-infract cortex compared with low-frequency (1-Hz) repetitive transcranial magnetic stimulation. The magnetic resonance spectroscopy data showed that low- and high-frequency repetitive transcranial magnetic stimulation reduced neuronal injury and maintained excitation/inhibition balance. However, 5-Hz repetitive transcranial magnetic stimulation showed more significant regulation of excitatory and inhibitory neurotransmitters after stroke than 1-Hz repetitive transcranial magnetic stimulation. These results indicated that high-frequency repetitive transcranial magnetic stimulation could more effectively promote neurovascular remodeling after stroke, and specific repetitive transcranial magnetic stimulation frequencies might be used to selectively regulate the neurovascular unit.
Collapse
Affiliation(s)
- Li Liu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ming Ding
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Junfa Wu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yuwen Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Qianfeng Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Nianhong Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Lu Luo
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Kewei Yu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yunhui Fan
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jingjun Zhang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yi Wu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiao Xiao
- Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Qun Zhang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
10
|
Boyle BR, Berghella AP, Blanco-Suarez E. Astrocyte Regulation of Neuronal Function and Survival in Stroke Pathophysiology. ADVANCES IN NEUROBIOLOGY 2024; 39:233-267. [PMID: 39190078 DOI: 10.1007/978-3-031-64839-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
The interactions between astrocytes and neurons in the context of stroke play crucial roles in the disease's progression and eventual outcomes. After a stroke, astrocytes undergo significant changes in their morphology, molecular profile, and function, together termed reactive astrogliosis. Many of these changes modulate how astrocytes relate to neurons, inducing mechanisms both beneficial and detrimental to stroke recovery. For example, excessive glutamate release and astrocytic malfunction contribute to excitotoxicity in stroke, eventually causing neuronal death. Astrocytes also provide essential metabolic support and neurotrophic signals to neurons after stroke, ensuring homeostatic stability and promoting neuronal survival. Furthermore, several astrocyte-secreted molecules regulate synaptic plasticity in response to stroke, allowing for the rewiring of neural circuits to compensate for damaged areas. In this chapter, we highlight the current understanding of the interactions between astrocytes and neurons in response to stroke, explaining the varied mechanisms contributing to injury progression and the potential implications for future therapeutic interventions.
Collapse
Affiliation(s)
- Bridget R Boyle
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrea P Berghella
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Elena Blanco-Suarez
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Neurological Surgery, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Chu MC, Mao WC, Wu HF, Chang YC, Lu TI, Lee CW, Chung YJ, Hsieh TH, Chang HS, Chen YF, Lin CH, Tang CW, Lin HC. Transient plasticity response is regulated by histone deacetylase inhibitor in oxygen-glucose deprivation condition. Pharmacol Rep 2023; 75:1200-1210. [PMID: 37695500 DOI: 10.1007/s43440-023-00525-w] [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: 06/01/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND The pathological form of synaptic plasticity, ischemic long-term potentiation (iLTP), induced by oxygen and glucose deprivation (OGD), is implicated in the acute phase of stroke with the potentiation of N-methyl-D-aspartate receptor (NMDAR). While there has been widespread attention on the excitatory system, a recent study reported that γ-aminobutyric acid (GABA)ergic system is also involved in iLTP. Valproic acid (VPA), a histone deacetylase inhibitor, protects against ischemic damage. However, whether VPA regulates early phase plasticity in ischemic stroke remains unknown. The present study aims to investigate the potential role and mechanism of VPA in ischemic stroke. METHODS A brief exposure of OGD on the hippocampal slices and the induction of photothrombotic ischemia (PTI) were used as ex vivo and in vivo models of ischemic stroke, respectively. RESULTS Using extracellular recordings, iLTP was induced in the hippocampal Schaffer collateral pathway following OGD exposure. VPA treatment abolished hippocampal iLTP via GABAA receptor enhancement and extracellular signal-regulated kinase (ERK) phosphorylation. Administration of VPA reduced brain infarct volume and motor dysfunction in mice with PTI. Moreover, VPA protected against ischemic injury by upregulating the GABAergic system and ERK phosphorylation, as well as by reducing of matrix metalloproteinase in a PTI-induced ischemic stroke model. CONCLUSIONS Together, this study revealed the protection of VPA in ex vivo OGD-induced pathological form of neuroplasticity and in vivo PTI-induced brain damage and motor dysfunction through rescuing GABAergic deficiency and the pathological hallmarks of ischemia.
Collapse
Affiliation(s)
- Ming-Chia Chu
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Chang Mao
- Department of Psychiatry, Cheng-Hsin General Hospital, Taipei, Taiwan
| | - Han-Fang Wu
- Department of Optometry, MacKay Medical College, New Taipei City, Taiwan
| | - Yun-Chi Chang
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ting-I Lu
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chi-Wei Lee
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yueh-Jung Chung
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tsung-Han Hsieh
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsun-Shuo Chang
- School of Pharmacy, College of Pharmacy, Kaoshiung Medical University, Kaoshiung, Taiwan
| | - Yih-Fung Chen
- School of Pharmacy, College of Pharmacy, Kaoshiung Medical University, Kaoshiung, Taiwan
- Graduate Institute of Natural Products, College of Pharmacy, Kaoshiung Medical University, Kaoshiung, Taiwan
| | - Chia-Hsien Lin
- Department of Health Industry Management, Kainan University, Taoyuan, Taiwan
| | - Chih-Wei Tang
- Department of Neurology, Far Eastern Memorial Hospital, New Taipei City, Taiwan.
| | - Hui-Ching Lin
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institute, Taipei, Taiwan.
| |
Collapse
|
12
|
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
|
13
|
Balena T, Lillis K, Rahmati N, Bahari F, Dzhala V, Berdichevsky E, Staley K. A Dynamic Balance between Neuronal Death and Clearance in an in Vitro Model of Acute Brain Injury. J Neurosci 2023; 43:6084-6107. [PMID: 37527922 PMCID: PMC10451151 DOI: 10.1523/jneurosci.0436-23.2023] [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: 03/09/2023] [Revised: 06/15/2023] [Accepted: 07/20/2023] [Indexed: 08/03/2023] Open
Abstract
In in vitro models of acute brain injury, neuronal death may overwhelm the capacity for microglial phagocytosis, creating a queue of dying neurons awaiting clearance. Neurons undergoing programmed cell death are in this queue, and are the most visible and frequently quantified measure of neuronal death after injury. However, the size of this queue should be equally sensitive to changes in neuronal death and the rate of phagocytosis. Using rodent organotypic hippocampal slice cultures as a model of acute perinatal brain injury, serial imaging demonstrated that the capacity for microglial phagocytosis of dying neurons was overwhelmed for 2 weeks. Altering phagocytosis rates (e.g., by changing the number of microglia) dramatically changed the number of visibly dying neurons. Similar effects were generated when the visibility of dying neurons was altered by changing the membrane permeability for stains that label dying neurons. Canonically neuroprotective interventions, such as seizure blockade, and neurotoxic maneuvers, such as perinatal ethanol exposure, were mediated by effects on microglial activity and the membrane permeability of neurons undergoing programmed cell death. These canonically neuroprotective and neurotoxic interventions had either no or opposing effects on healthy surviving neurons identified by the ongoing expression of transgenic fluorescent proteins.SIGNIFICANCE STATEMENT In in vitro models of acute brain injury, microglial phagocytosis is overwhelmed by the number of dying cells. Under these conditions, the assumptions on which assays for neuroprotective and neurotoxic effects are based are no longer valid. Thus, longitudinal assays of healthy cells, such as serial assessment of the fluorescence emission of transgenically expressed proteins, provide more accurate estimates of cell death than do single-time point anatomic or biochemical assays of the number of dying neurons. More accurate estimates of death rates in vitro will increase the translatability of preclinical studies of neuroprotection and neurotoxicity.
Collapse
Affiliation(s)
- Trevor Balena
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Kyle Lillis
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Negah Rahmati
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Fatemeh Bahari
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Volodymyr Dzhala
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Eugene Berdichevsky
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, Pennsylvania 18015
| | - Kevin Staley
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| |
Collapse
|
14
|
Seo HW, Ha TY, Ko G, Jang A, Choi JW, Lee DH, Chang KA. Scutellaria baicalensis Attenuated Neurological Impairment by Regulating Programmed Cell Death Pathway in Ischemic Stroke Mice. Cells 2023; 12:2133. [PMID: 37681864 PMCID: PMC10486384 DOI: 10.3390/cells12172133] [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: 07/21/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 09/09/2023] Open
Abstract
Stroke is a major global health problem that causes significant mortality and long-term disability. Post-stroke neurological impairment is a complication that is often underestimated with the risk of persistent neurological deficits. Although traditional Chinese medicines have a long history of being used for stroke, their scientific efficacy remains unclear. Scutellaria baicalensis, an herbal component known for its anti-inflammatory and antioxidant properties, has traditionally been used to treat brain disorders. This study investigated the therapeutic effects of the Scutellaria baicalensis extraction (SB) during the acute stage of ischemic stroke using photothrombotic (PTB)-induced and transient middle cerebral artery occlusion (tMCAO) model mice. We found that SB mitigated ischemic brain injury, as evidenced by a significant reduction in the modified neurological severity score in the acute stage of PTB and both the acute and chronic stages of tMCAO. Furthermore, we elucidated the regulatory role of SB in the necroptosis and pyroptosis pathways during the acute stage of stroke, underscoring its protective effects. Behavioral assessments demonstrated the effectiveness of SB in ameliorating motor dysfunction and cognitive impairment compared to the group receiving the vehicle. Our findings highlight the potential of SB as a promising therapeutic candidate for stroke. SB was found to help modulate the programmed cell death pathways, promote neuroprotection, and facilitate functional recovery.
Collapse
Affiliation(s)
- Ho-won Seo
- Department of Health Science and Technology, Gachon Advanced Institute for Health Sciences & Technology, Gachon University, Incheon 21999, Republic of Korea; (H.-w.S.); (G.K.)
| | - Tae-Young Ha
- Neuroscience Research Institute, Gachon University, Incheon 21565, Republic of Korea;
| | - Geon Ko
- Department of Health Science and Technology, Gachon Advanced Institute for Health Sciences & Technology, Gachon University, Incheon 21999, Republic of Korea; (H.-w.S.); (G.K.)
| | - Aram Jang
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Gyeonggi-do, Republic of Korea;
| | - Ji-Woong Choi
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, Republic of Korea;
| | - Dong-hun Lee
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Gyeonggi-do, Republic of Korea;
| | - Keun-A Chang
- Department of Health Science and Technology, Gachon Advanced Institute for Health Sciences & Technology, Gachon University, Incheon 21999, Republic of Korea; (H.-w.S.); (G.K.)
- Neuroscience Research Institute, Gachon University, Incheon 21565, Republic of Korea;
- Department of Pharmacology, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
15
|
Rivera DA, Schaffer CB. Quasi-analytic solution for real-time multi-exposure speckle imaging of tissue perfusion. BIOMEDICAL OPTICS EXPRESS 2023; 14:3950-3967. [PMID: 37799691 PMCID: PMC10549738 DOI: 10.1364/boe.493821] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/23/2023] [Accepted: 06/25/2023] [Indexed: 10/07/2023]
Abstract
Laser speckle contrast imaging (LSCI) is a widefield imaging technique that enables high spatiotemporal resolution measurement of blood flow. Laser coherence, optical aberrations, and static scattering effects restrict LSCI to relative and qualitative measurements. Multi-exposure speckle imaging (MESI) is a quantitative extension of LSCI that accounts for these factors but has been limited to post-acquisition analysis due to long data processing times. Here we propose and test a real-time quasi-analytic solution to fitting MESI data, using both simulated and real-world data from a mouse model of photothrombotic stroke. This rapid estimation of multi-exposure imaging (REMI) enables processing of full-frame MESI images at up to 8 Hz with negligible errors relative to time-intensive least-squares methods. REMI opens the door to real-time, quantitative measures of perfusion change using simple optical systems.
Collapse
Affiliation(s)
- Daniel A Rivera
- Nance E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Chris B Schaffer
- Nance E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| |
Collapse
|
16
|
Shabani Z, Farhoudi M, Rahbarghazi R, Karimipour M, Mehrad H. Cellular, histological, and behavioral pathological alterations associated with the mouse model of photothrombotic ischemic stroke. J Chem Neuroanat 2023; 130:102261. [PMID: 36967096 DOI: 10.1016/j.jchemneu.2023.102261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/13/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND Photothrombotic (PT) stroke model is a reliable method to induce ischemic stroke in the target site using the excitation of photosensitive agents such as Rose Bengal (RB) dye after light illumination. Here, we performed a PT-induced brain ischemic model using a green laser and photosensitive agent RB and confirmed its efficiency through cellular, histological, and neurobehavioral approaches. METHODS Mice were randomly allocated into RB; Laser irradiation; and RB + Laser irradiation groups. Mice were exposed to a green laser at a wavelength of 532 nm and intensity of 150 mW in a mouse model after injection of RB under stereotactic surgery. The pattern of Hemorrhagic and ischemic changes were evaluated throughout the study. The volume of the lesion site was calculated using unbiased stereological methods. For investigation of neurogenesis, we performed double - (BrdU/NeuN) immunofluorescence (IF) staining on day 28 following the last- BrdU injection. To assess the effect and quality of ischemic stroke on neurological behavior, the Modified neurological severity score (mNSS) test was done on days 1, 7, 14, and 28 days after stroke induction. RESULTS Laser irradiation plus RB induced hemorrhagic tissue and pale ischemic changes over the 5 days. In the next few days, microscopic staining revealed neural tissue degeneration, demarcated necrotic site, and neuronal injury. BrdU staining showed a significant number of proliferating cells in the periphery of the lesion site in the Laser irradiation plus RB group compared to the group (p < 0.05) while the percent of NeuN+ cells per BrdU- positive cells was reduced. Also, prominent astrogliosis was observed in the periphery of irradiated sites on day 28. Neurological deficits were detected in mice from Laser irradiation plus the RB group. No histological or functional deficits were detected in RB and Laser irradiation groups. CONCLUSIONS Taken together, our study showed cellular and histologic pathological changes which are associated with the PT induction model. Our findings indicated that the undesirable microenvironment and inflammatory conditions could affect neurogenesis concomitantly with functional deficits. Moreover, this research showed that this model is a focal, reproducible, noninvasive and accessible stroke model with a distinctive demarcation similar to human stroke conditions.
Collapse
|
17
|
Sokolowski JD, Soldozy S, Sharifi KA, Norat P, Kearns KN, Liu L, Williams AM, Yağmurlu K, Mastorakos P, Miller GW, Kalani MYS, Park MS, Kellogg RT, Tvrdik P. Preclinical models of middle cerebral artery occlusion: new imaging approaches to a classic technique. Front Neurol 2023; 14:1170675. [PMID: 37409019 PMCID: PMC10318149 DOI: 10.3389/fneur.2023.1170675] [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: 02/21/2023] [Accepted: 05/30/2023] [Indexed: 07/07/2023] Open
Abstract
Stroke remains a major burden on patients, families, and healthcare professionals, despite major advances in prevention, acute treatment, and rehabilitation. Preclinical basic research can help to better define mechanisms contributing to stroke pathology, and identify therapeutic interventions that can decrease ischemic injury and improve outcomes. Animal models play an essential role in this process, and mouse models are particularly well-suited due to their genetic accessibility and relatively low cost. Here, we review the focal cerebral ischemia models with an emphasis on the middle cerebral artery occlusion technique, a "gold standard" in surgical ischemic stroke models. Also, we highlight several histologic, genetic, and in vivo imaging approaches, including mouse stroke MRI techniques, that have the potential to enhance the rigor of preclinical stroke evaluation. Together, these efforts will pave the way for clinical interventions that can mitigate the negative impact of this devastating disease.
Collapse
Affiliation(s)
- Jennifer D. Sokolowski
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
| | - Sauson Soldozy
- Department of Neurological Surgery, Westchester Medical Center, Valhalla, NY, United States
| | - Khadijeh A. Sharifi
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
- Department of Neuroscience, University of Virginia, Charlottesville, VA, United States
| | - Pedro Norat
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
| | - Kathryn N. Kearns
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
| | - Lei Liu
- Department of Neurological Surgery and Neuroscience, Northwestern University, Chicago, IL, United States
| | - Ashley M. Williams
- School of Medicine, Morsani College of Medicine, Tampa, FL, United States
| | - Kaan Yağmurlu
- Department of Neurological Surgery, University of Tennessee, Memphis, TN, United States
| | - Panagiotis Mastorakos
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, United States
| | - G. Wilson Miller
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, United States
| | - M. Yashar S. Kalani
- Department of Neurological Surgery, St. John's Neuroscience Institute, Tulsa, OK, United States
| | - Min S. Park
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
| | - Ryan T. Kellogg
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
| | - Petr Tvrdik
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
- Department of Neuroscience, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
18
|
Menozzi L, Del Águila Á, Vu T, Ma C, Yang W, Yao J. Integrated Photoacoustic, Ultrasound, and Angiographic Tomography (PAUSAT) for NonInvasive Whole-Brain Imaging of Ischemic Stroke. J Vis Exp 2023:10.3791/65319. [PMID: 37335115 PMCID: PMC10411115 DOI: 10.3791/65319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2023] Open
Abstract
Presented here is an experimental ischemic stroke study using our newly developed noninvasive imaging system that integrates three acoustic-based imaging technologies: photoacoustic, ultrasound, and angiographic tomography (PAUSAT). Combining these three modalities helps acquire multi-spectral photoacoustic tomography (PAT) of the brain blood oxygenation, high-frequency ultrasound imaging of the brain tissue, and acoustic angiography of the cerebral blood perfusion. The multi-modal imaging platform allows the study of cerebral perfusion and oxygenation changes in the whole mouse brain after stroke. Two commonly used ischemic stroke models were evaluated: the permanent middle cerebral artery occlusion (pMCAO) model and the photothrombotic (PT) model. PAUSAT was used to image the same mouse brains before and after a stroke and quantitatively analyze both stroke models. This imaging system was able to clearly show the brain vascular changes after ischemic stroke, including significantly reduced blood perfusion and oxygenation in the stroke infarct region (ipsilateral) compared to the uninjured tissue (contralateral). The results were confirmed by both laser speckle contrast imaging and triphenyltetrazolium chloride (TTC) staining. Furthermore, stroke infarct volume in both stroke models was measured and validated by TTC staining as the ground truth. Through this study, we have demonstrated that PAUSAT can be a powerful tool in noninvasive and longitudinal preclinical studies of ischemic stroke.
Collapse
Affiliation(s)
- Luca Menozzi
- Department of Biomedical Engineering, Duke University
| | - Ángela Del Águila
- Multidisciplinary Brain Protection Program, Department of Anesthesiology, Duke University School of Medicine
| | - Tri Vu
- Department of Biomedical Engineering, Duke University
| | - Chenshuo Ma
- Department of Biomedical Engineering, Duke University
| | - Wei Yang
- Multidisciplinary Brain Protection Program, Department of Anesthesiology, Duke University School of Medicine;
| | - Junjie Yao
- Department of Biomedical Engineering, Duke University;
| |
Collapse
|
19
|
Mosberger AC, Sibener LJ, Chen TX, Rodrigues H, Hormigo R, Ingram JN, Athalye VR, Tabachnik T, Wolpert DM, Murray JM, Costa RM. Exploration biases how forelimb reaches to a spatial target are learned. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.08.539291. [PMID: 37214823 PMCID: PMC10197595 DOI: 10.1101/2023.05.08.539291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The brain can learn to generate actions, such as reaching to a target, using different movement strategies. Understanding how different variables bias which strategies are learned to produce such a reach is important for our understanding of the neural bases of movement. Here we introduce a novel spatial forelimb target task in which perched head-fixed mice learn to reach to a circular target area from a set start position using a joystick. These reaches can be achieved by learning to move into a specific direction or to a specific endpoint location. We find that mice gradually learn to successfully reach the covert target. With time, they refine their initially exploratory complex joystick trajectories into controlled targeted reaches. The execution of these controlled reaches depends on the sensorimotor cortex. Using a probe test with shifting start positions, we show that individual mice learned to use strategies biased to either direction or endpoint-based movements. The degree of endpoint learning bias was correlated with the spatial directional variability with which the workspace was explored early in training. Furthermore, we demonstrate that reinforcement learning model agents exhibit a similar correlation between directional variability during training and learned strategy. These results provide evidence that individual exploratory behavior during training biases the control strategies that mice use to perform forelimb covert target reaches.
Collapse
|
20
|
Choi SG, Shin J, Lee KY, Park H, Kim SI, Yi YY, Kim DW, Song HJ, Shin HJ. PINK1 siRNA-loaded poly(lactic-co-glycolic acid) nanoparticles provide neuroprotection in a mouse model of photothrombosis-induced ischemic stroke. Glia 2023; 71:1294-1310. [PMID: 36655313 DOI: 10.1002/glia.24339] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/14/2022] [Accepted: 01/05/2023] [Indexed: 01/20/2023]
Abstract
PTEN-induced kinase 1 (PINK1) is a well-known critical marker in the pathway for mitophagy regulation as well as mitochondrial dysfunction. Evidence suggests that mitochondrial dynamics and mitophagy flux play an important role in the development of brain damage from stroke pathogenesis. In this study, we propose a treatment strategy using nanoparticles that can control PINK1. We used a murine photothrombotic ischemic stroke (PTS) model in which clogging of blood vessels is induced with Rose Bengal (RB) to cause brain damage. We targeted PINK1 with poly(lactic-co-glycolic acid) (PLGA)-based nanoparticles loaded with PINK1 siRNA (PINK1 NPs). After characterizing siRNA loading in the nanoparticles, we assessed the efficacy of PINK1 NPs in mice with PTS using immunohistochemistry, 1% 2,3,5-triphenyltetrazolium chloride staining, measurement of motor dysfunction, and Western blot. PINK1 was highly expressed in microglia 24 h after PTS induction. PINK1 siRNA treatment increased phagocytic activity, migration, and expression of an anti-inflammatory state in microglia. In addition, the PLGA nanoparticles were selectively taken up by microglia and specifically regulated PINK1 expression in those cells. Treatment with PINK1 NPs prior to stroke induction reduced expression of mitophagy-inducing factors, infarct volume, and motor dysfunction in mice with photothrombotic ischemia. Experiments with PINK1-knockout mice and microglia depletion with PLX3397 confirmed a decrease in stroke-induced infarct volume and behavioral dysfunction. Application of nanoparticles for PINK1 inhibition attenuates RB-induced photothrombotic ischemic injury by inhibiting microglia responses, suggesting that a nanomedical approach targeting the PINK1 pathway may provide a therapeutic avenue for stroke treatment.
Collapse
Affiliation(s)
- Seung Gyu Choi
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Juhee Shin
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Ka Young Lee
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Hyewon Park
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Song I Kim
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Yoon Young Yi
- Department of Pediatrics, College of Medicine, Hallym University and Gangdong Sacred Heart Hospital, Seoul, Republic of Korea
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Hee-Jung Song
- Department of Neurology, Chungnam National University Sejong Hospital and College of Medicine, Republic of Korea
| | - Hyo Jung Shin
- Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
21
|
Brezgunova AA, Andrianova NV, Popkov VA, Tkachev SY, Manskikh VN, Pevzner IB, Zorova LD, Timashev PS, Silachev DN, Zorov DB, Plotnikov EY. New experimental model of kidney injury: Photothrombosis-induced kidney ischemia. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166622. [PMID: 36526237 DOI: 10.1016/j.bbadis.2022.166622] [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: 07/22/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022]
Abstract
Acute kidney injury (AKI) is a frequent pathology with a high mortality rate after even a single AKI episode and a great risk of chronic kidney disease (CKD) development. To get insight into mechanisms of the AKI pathogenesis, there is a need to develop diverse experimental models of the disease. Photothrombosis is a widely used method for inducing ischemia in the brain. In this study, for the first time, we described photothrombosis-induced kidney ischemia as an appropriate model of AKI and obtained comprehensive characteristics of the photothrombotic lesion using micro-computed tomography (micro-CT) and histological techniques. In the ischemic area, we observed destruction of tubules, the loss of brush border and nuclei, connective tissue fibers disorganization, leukocyte infiltration, and hyaline casts formation. In kidney tissue and urine, we revealed increased levels in markers of proliferation and injury. The explicit long-term consequence of photothrombosis-induced kidney ischemia was renal fibrosis. Thus, we establish a new low invasive experimental model of AKI, which provides a reproducible local ischemic injury lesion. We propose our model of photothrombosis-induced kidney ischemia as a useful approach for investigating AKI pathogenesis, studying the mechanisms of kidney regeneration, and development of therapy against AKI and CKD.
Collapse
Affiliation(s)
- Anna A Brezgunova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia; A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Nadezda V Andrianova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Vasily A Popkov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, Russia
| | - Sergey Y Tkachev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Vasily N Manskikh
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Irina B Pevzner
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, Russia
| | - Ljubava D Zorova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, Russia
| | - Peter S Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Denis N Silachev
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, Russia
| | - Dmitry B Zorov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, Russia.
| | - Egor Y Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia; V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow, Russia.
| |
Collapse
|
22
|
Balena T, Lillis K, Rahmati N, Bahari F, Dzhala V, Berdichevsky E, Staley K. A dynamic balance between neuronal death and clearance after acute brain injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.14.528332. [PMID: 36824708 PMCID: PMC9948967 DOI: 10.1101/2023.02.14.528332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
After acute brain injury, neuronal apoptosis may overwhelm the capacity for microglial phagocytosis, creating a queue of dying neurons awaiting clearance. The size of this queue should be equally sensitive to changes in neuronal death and the rate of phagocytosis. Using rodent organotypic hippocampal slice cultures as a model of acute perinatal brain injury, serial imaging demonstrated that the capacity for microglial phagocytosis of dying neurons was overwhelmed for two weeks. Altering phagocytosis rates, e.g. by changing the number of microglia, dramatically changed the number of visibly dying neurons. Similar effects were generated when the visibility of dying neurons was altered by changing the membrane permeability for vital stains. Canonically neuroprotective interventions such as seizure blockade and neurotoxic maneuvers such as perinatal ethanol exposure were mediated by effects on microglial activity and the membrane permeability of apoptotic neurons, and had either no or opposing effects on healthy surviving neurons. Significance After acute brain injury, microglial phagocytosis is overwhelmed by the number of dying cells. Under these conditions, the assumptions on which assays for neuroprotective and neurotoxic effects are based are no longer valid. Thus longitudinal assays of healthy cells, such as assessment of the fluorescence emission of transgenically-expressed proteins, provide more accurate estimates of cell death than do single-time-point anatomical or biochemical assays. More accurate estimates of death rates will increase the translatability of preclinical studies of neuroprotection and neurotoxicity.
Collapse
|
23
|
Conti E, Carlini N, Piccardi B, Allegra Mascaro AL, Pavone FS. Photothrombotic Middle Cerebral Artery Occlusion in Mice: A Novel Model of Ischemic Stroke. eNeuro 2023; 10:ENEURO.0244-22.2022. [PMID: 36650068 PMCID: PMC9910575 DOI: 10.1523/eneuro.0244-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/25/2022] [Accepted: 11/06/2022] [Indexed: 01/19/2023] Open
Abstract
Stroke is one of the main causes of death and disability worldwide. Over the past decades, several animal models of focal cerebral ischemia have been developed allowing to investigate pathophysiological mechanisms underlying stroke progression. Despite intense preclinical research efforts, the need for noninvasive mouse models of vascular occlusion targeting the middle cerebral artery yet avoiding mechanical intervention is still pressing. Here, by applying the photothrombotic stroke model to the distal branch of the middle cerebral artery, we developed a novel strategy to induce a targeted occlusion of a large blood vessel in mice. This approach induces unilateral damage encompassing most of the dorsal cortex from the motor up to the visual regions 1 week after stroke. Pronounced limb dystonia one day after the damage is partially recovered after one week. Furthermore, we observe the insurgence of blood vessel leakage and edema formation in the peri-infarct area. Finally, this model elicits a notable inflammatory response revealed as a strong increase in astrocyte density and morphologic complexity in the perilesional region of the cortex compared with both other regions of the ipsilesional and contralesional hemispheres, and in sham-operated mice. To conclude, the stroke model we developed induces in mice the light-mediated occlusion of one of the main targets of human ischemic stroke, the middle cerebral artery, free from the limitations of commonly used preclinical models.
Collapse
Affiliation(s)
- Emilia Conti
- Neuroscience Institute, National Research Council, 56124 Pisa, Italy
- European Laboratory for Non-Linear Spectroscopy, 50019 Sesto Fiorentino, Italy
- Translational Research on Stroke (TREES) Working Group, Florence, Italy
| | - Noemi Carlini
- Neuroscience Institute, National Research Council, 56124 Pisa, Italy
- European Laboratory for Non-Linear Spectroscopy, 50019 Sesto Fiorentino, Italy
| | - Benedetta Piccardi
- Neurofarba Department, University of Florence, 50139 Florence, Italy
- Translational Research on Stroke (TREES) Working Group, Florence, Italy
| | - Anna Letizia Allegra Mascaro
- Neuroscience Institute, National Research Council, 56124 Pisa, Italy
- European Laboratory for Non-Linear Spectroscopy, 50019 Sesto Fiorentino, Italy
- Translational Research on Stroke (TREES) Working Group, Florence, Italy
| | - Francesco Saverio Pavone
- European Laboratory for Non-Linear Spectroscopy, 50019 Sesto Fiorentino, Italy
- Department of Physics and Astronomy, University of Florence, 50019 Sesto Fiorentino, Italy
- National Institute of Optics, National Research Council, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
24
|
Ke X, Deng M, Wu Z, Yu H, Yu D, Li H, Lu Y, Shu K, Pei L. miR-34b-3p Inhibition of eIF4E Causes Post-stroke Depression in Adult Mice. Neurosci Bull 2023; 39:194-212. [PMID: 35802246 PMCID: PMC9905405 DOI: 10.1007/s12264-022-00898-7] [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: 02/23/2022] [Accepted: 05/14/2022] [Indexed: 11/30/2022] Open
Abstract
Post-stroke depression (PSD) is a serious and common complication of stroke, which seriously affects the rehabilitation of stroke patients. To date, the pathogenesis of PSD is unclear and effective treatments remain unavailable. Here, we established a mouse model of PSD through photothrombosis-induced focal ischemia. By using a combination of brain imaging, transcriptome sequencing, and bioinformatics analysis, we found that the hippocampus of PSD mice had a significantly lower metabolic level than other brain regions. RNA sequencing revealed a significant reduction of miR34b-3p, which was expressed in hippocampal neurons and inhibited the translation of eukaryotic translation initiation factor 4E (eIF4E). Furthermore, silencing eIF4E inactivated microglia, inhibited neuroinflammation, and abolished the depression-like behaviors in PSD mice. Together, our data demonstrated that insufficient miR34b-3p after stroke cannot inhibit eIF4E translation, which causes PSD by the activation of microglia in the hippocampus. Therefore, miR34b-3p and eIF4E may serve as potential therapeutic targets for the treatment of PSD.
Collapse
Affiliation(s)
- Xiao Ke
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Manfei Deng
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhuoze Wu
- Department of Pathophysiology, Basic Medical School, North Sichuan Medical College, Nanchong, 637100, China
| | - Hongyan Yu
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dian Yu
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hao Li
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Youming Lu
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lei Pei
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
25
|
Gao D, Li Y, Wu Y, Liu Y, Hu D, Liang S, Liao J, Pan M, Zhang P, Li K, Liu X, Zheng H, Sheng Z. Albumin-Consolidated AIEgens for Boosting Glioma and Cerebrovascular NIR-II Fluorescence Imaging. ACS APPLIED MATERIALS & INTERFACES 2023; 15:3-13. [PMID: 34995067 DOI: 10.1021/acsami.1c22700] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The application of an exogenous polymer matrix to construct aggregation-induced emission (AIE) nanoprobes promotes the utility of AIE luminogens (AIEgens) in diagnosing brain diseases. However, the limited fluorescence (FL) and low active-targeting abilities of AIE-based nanoprobes impede their imaging application. Here, we employed endogenous albumin as an effective matrix to encapsulate AIEgens to enhance FL quantum yield (QY) and active-targeting ability. The albumin-consolidated strategy effectively inhibited the intramolecular vibration of AIEgens and enhanced endocytosis mediated by the gp60 receptor. The QYs of three kinds of albumin-based AIE nanoprobes with FL emissions ranging from the visible (400-650 nm) to the second near-infrared (NIR-II, 1000-1700 nm) region was at least 10% higher, and the tumor-targeting efficiency was ∼25% higher, compared with those of nanoprobes constructed by the exogenous polymer. Albumin-based AIE nanoprobes have achieved active-targeting NIR-II imaging of brain tumors and cerebrovascular imaging with a high signal-to-background ratio (SBR, ∼90) and high resolution (∼70 μm) in mouse models. Therefore, the albumin-based AIE nanoprobes will enable FL imaging-guided surgery of brain tumors and cerebral ischemia, which will improve surgical efficacy to prevent recurrence and side effects.
Collapse
Affiliation(s)
- Duyang Gao
- Paul C. Lauterbur Research Center for Biomedical Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Yaxi Li
- Department of Biomedical Engineering, SUSTech Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yayun Wu
- Paul C. Lauterbur Research Center for Biomedical Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Yu Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Dehong Hu
- Paul C. Lauterbur Research Center for Biomedical Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Simin Liang
- Department of Ultrasonography, Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen 518034, China
| | - Jiuling Liao
- Paul C. Lauterbur Research Center for Biomedical Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Min Pan
- Department of Ultrasonography, Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen 518034, China
| | - Pengfei Zhang
- Paul C. Lauterbur Research Center for Biomedical Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Kai Li
- Department of Biomedical Engineering, SUSTech Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xin Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| | - Zonghai Sheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, CAS Key Laboratory of Health Informatics, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China
| |
Collapse
|
26
|
Venkat P, Gao H, Findeis EL, Chen Z, Zacharek A, Landschoot-Ward J, Powell B, Lu M, Liu Z, Zhang Z, Chopp M. Therapeutic effects of CD133 + Exosomes on liver function after stroke in type 2 diabetic mice. Front Neurosci 2023; 17:1061485. [PMID: 36968490 PMCID: PMC10033607 DOI: 10.3389/fnins.2023.1061485] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/21/2023] [Indexed: 03/29/2023] Open
Abstract
Background and purpose Non-alcoholic fatty liver disease (NAFLD) is known to adversely affect stroke recovery. However, few studies investigate how stroke elicits liver dysfunction, particularly, how stroke in type 2 diabetes mellitus (T2DM) exacerbates progression of NAFLD. In this study, we test whether exosomes harvested from human umbilical cord blood (HUCBC) derived CD133 + cells (CD133 + Exo) improves neuro-cognitive outcome as well as reduces liver dysfunction in T2DM female mice. Methods Female, adult non-DM and T2DM mice subjected to stroke presence or absence were considered. T2DM-stroke mice were randomly assigned to receive PBS or Exosome treatment group. CD133 + Exo (20 μg/200 μl PBS, i.v.) was administered once at 3 days after stroke. Evaluation of neurological (mNSS, adhesive removal test) and cognitive function [novel object recognition (NOR) test, odor test] was performed. Mice were sacrificed at 28 days after stroke and brain, liver, and serum were harvested. Results Stroke induces severe and significant short-term and long-term neurological and cognitive deficits which were worse in T2DM mice compared to non-DM mice. CD133 + Exo treatment of T2DM-stroke mice significantly improved neurological function and cognitive outcome indicated by improved discrimination index in the NOR and odor tests compared to control T2DM-stroke mice. CD133 + Exo treatment of T2DM stroke significantly increased vascular and white matter/axon remodeling in the ischemic brain compared to T2DM-stroke mice. However, there were no differences in the lesion volume between non-DM stroke, T2DM-stroke and CD133 + Exo treated T2DM-stroke mice. In T2DM mice, stroke induced earlier and higher TLR4, NLRP3, and cytokine expression (SAA, IL1β, IL6, TNFα) in the liver compared to heart and kidney, as measured by Western blot. T2DM-stroke mice exhibited worse NAFLD progression with increased liver steatosis, hepatocellular ballooning, fibrosis, serum ALT activity, and higher NAFLD Activity Score compared to T2DM mice and non-DM-stroke mice, while CD133 + Exo treatment significantly attenuated the progression of NAFLD in T2DM stroke mice. Conclusion Treatment of female T2DM-stroke mice with CD133 + Exo significantly reduces the progression of NAFLD/NASH and improves neurological and cognitive function compared to control T2DM-stroke mice.
Collapse
Affiliation(s)
- Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
- *Correspondence: Poornima Venkat,
| | - Huanjia Gao
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | | | - Zhili Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | | | - Brianna Powell
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Mei Lu
- Department of Public Health Sciences, Henry Ford Hospital, Detroit, MI, United States
| | - Zhongwu Liu
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Zhenggang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
- Department of Physics, Oakland University, Rochester, MI, United States
| |
Collapse
|
27
|
Semenov DG, Belyakov AV, Rybnikova EA. Experimental Modeling of Damaging and Protective Hypoxia of the Mammalian Brain. J EVOL BIOCHEM PHYS+ 2022; 58:2021-2034. [PMID: 36573160 PMCID: PMC9773672 DOI: 10.1134/s0022093022060291] [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: 09/02/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 12/24/2022]
Abstract
Currently, there is a new surge of interest in the problem of hypoxia, almost lost in recent decades. Due to the fact that the circle of competent specialists in this field has significantly narrowed, it is necessary to carry out an intensive exchange of knowledge. In order to inform a wide range of interested researchers and doctors, this review summarizes the current understanding of hypoxia, its pathogenic and adaptogenic consequences, as well as key physiological and molecular mechanisms that implement the response to hypoxia at various levels-from cellular to organismic. The review presents a modern classification of forms of hypoxia, the understanding of which is necessary for the formation of a scientifically based approach to experimental modeling of hypoxic states. An analysis of the literature covering the history and current level of hypoxia modeling in mammals and human experiments, including methods for creating moderate hypoxia used to increase the resistance of the nervous system to severe forms of hypoxia and other extreme factors, is carried out. Special attention is paid to the discussion of the features and limitations of various approaches to the creation of hypoxia, as well as the disclosure of the potential for the practical application of moderate hypoxic effects in medicine.
Collapse
Affiliation(s)
- D G Semenov
- Pavlov Institute of Physiology of Russian Academy of Sciences, St. Petersburg, Russia
| | - A V Belyakov
- Pavlov Institute of Physiology of Russian Academy of Sciences, St. Petersburg, Russia
| | - E A Rybnikova
- Pavlov Institute of Physiology of Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
28
|
The Endothelial Glycocalyx and Retinal Hemodynamics. PATHOPHYSIOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY FOR PATHOPHYSIOLOGY 2022; 29:663-677. [PMID: 36548208 PMCID: PMC9785437 DOI: 10.3390/pathophysiology29040052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/20/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
PURPOSE Previous studies suggest that the endothelial glycocalyx adds to vascular resistance, inhibits thrombosis, and is critical for regulating homogeneous blood flow and ensuring uniform red blood cell (RBC) distribution. However, these functions and consequences of the glycocalyx have not been examined in the retina. We hypothesize that the endothelial glycocalyx is a critical regulator of retinal hemodynamics and perfusion and decreases the propensity for retinal thrombus formation. METHODS Hyaluronidase and heparinase, which are endothelial glycocalyx-degrading enzymes, were infused into mice. Fluorescein isothiocyanate-dextran (2000 kDa) was injected to measure lumen diameter, while RBC velocity and distribution were measured using fluorescently labeled RBCs. The diameters and velocities were used to calculate retinal blood flow and shear rates. Mean circulation time was calculated by measuring the difference between arteriolar and venular mean transit times. Rose Bengal dye was infused, followed by illumination with a green light to induce thrombosis. RESULTS The acute infusion of hyaluronidase and heparinase led to significant increases in both arteriolar (7%) and venular (16%) diameters in the retina, with a tendency towards increased arteriolar velocity. In addition, the degradation caused a significant decrease in the venular shear rate (14%). The enzyme infusion resulted in substantial increases in total retinal blood flow (26%) and retinal microhematocrit but no changes in the mean circulation time through the retina. We also observed an enhanced propensity for retinal thrombus formation with the removal of the glycocalyx. CONCLUSIONS Our data suggest that acute degradation of the glycocalyx can cause significant changes in retinal hemodynamics, with increases in vessel diameter, blood flow, microhematocrit, pro-thrombotic conditions, and decreases in venular shear rate.
Collapse
|
29
|
Weber RZ, Mulders G, Perron P, Tackenberg C, Rust R. Molecular and anatomical roadmap of stroke pathology in immunodeficient mice. Front Immunol 2022; 13:1080482. [PMID: 36569903 PMCID: PMC9785704 DOI: 10.3389/fimmu.2022.1080482] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
Background Stroke remains a leading cause of disability and death worldwide. It has become apparent that inflammation and immune mediators have a pre-dominant role in initial tissue damage and long-term recovery. Still, different immunosuppressed mouse models are necessary in stroke research e.g., to evaluate therapies using human cell grafts. Despite mounting evidence delineating the importance of inflammation in the stroke pathology, it is poorly described to what extent immune deficiency influences overall stroke outcome. Methods Here, we assessed the stroke pathology of popular genetic immunodeficient mouse models, i.e., NOD scid gamma (NSG) and recombination activating gene 2 (Rag2-/-) mice as well as pharmacologically immunosuppressed mice and compared them to immune competent, wildtype (WT) C57BL/6J mice three weeks after injury. We performed histology, gene expression, blood serum and behavioural analysis to identify the impact of immunosuppression on stroke progression. Results We detected changes in microglia activation/macrophage infiltration, scar-forming and vascular repair in immune-suppressed mice three weeks after injury. Transcriptomic analysis of stroked tissue revealed the strongest deviation from WT was observed in NSG mice affecting immunological and angiogenic pathways. Pharmacological immunosuppression resulted in the least variation in gene expression compared with the WT. These anatomical and genetic changes did not affect functional recovery in a time course of three weeks. To determine whether timing of immunosuppression is critical, we compared mice with acute and delayed pharmacological immunosuppression after stroke. Mice with delayed immunosuppression (7d) showed increased inflammatory and scarring responses compared to animals acutely treated with tacrolimus, thus more closely resembling WT pathology. Transplantation of human cells in the brains of immunosuppressed mice led to prolonged cell survival in all immunosuppressed mouse models, which was most consistent in NSG and Rag2-/- mice. Conclusions We detected distinct anatomical and molecular changes in the stroke pathology between individual immunosuppressed mouse models that should be considered when selecting an appropriate mouse model for stroke research.
Collapse
Affiliation(s)
- Rebecca Z. Weber
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland,Neuroscience Center Zurich, University of Zurich and Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Geertje Mulders
- Department of Health Sciences and Technology, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Patrick Perron
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
| | - Christian Tackenberg
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland,Neuroscience Center Zurich, University of Zurich and Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Ruslan Rust
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland,Neuroscience Center Zurich, University of Zurich and Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland,*Correspondence: Ruslan Rust,
| |
Collapse
|
30
|
Walter HL, Pikhovych A, Endepols H, Rotthues S, Bärmann J, Backes H, Hoehn M, Wiedermann D, Neumaier B, Fink GR, Rüger MA, Schroeter M. Transcranial-Direct-Current-Stimulation Accelerates Motor Recovery After Cortical Infarction in Mice: The Interplay of Structural Cellular Responses and Functional Recovery. Neurorehabil Neural Repair 2022; 36:701-714. [PMID: 36124996 DOI: 10.1177/15459683221124116] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Transcranial direct current stimulation (tDCS) promotes recovery after stroke in humans. The underlying mechanisms, however, remain to be elucidated. Animal models suggest tDCS effects on neuroinflammation, stem cell proliferation, neurogenesis, and neural plasticity. OBJECTIVE In a longitudinal study, we employed tDCS in the subacute and chronic phase after experimental focal cerebral ischemia in mice to explore the relationship between functional recovery and cellular processes. METHODS Mice received photothrombosis in the right motor cortex, verified by Magnetic Resonance Imaging. A composite neuroscore quantified subsequent functional deficits. Mice received tDCS daily: either 5 sessions from day 5 to 9, or 10 sessions with days 12 to 16 in addition. TDCS with anodal or cathodal polarity was compared to sham stimulation. Further imaging to assess proliferation and neuroinflammation was performed by immunohistochemistry at different time points and Positron Emission Tomography at the end of the observation time of 3 weeks. RESULTS Cathodal tDCS at 198 kC/m2 (220 A/m2) between days 5 and 9 accelerated functional recovery, increased neurogenesis, decreased microglial activation, and mitigated CD16/32-expression associated with M1-phenotype. Anodal tDCS exerted similar effects on neurogenesis and microglial polarization but not on recovery of function or microglial activation. TDCS on days 12 to 16 after stroke did not induce any further effects, suggesting that the therapeutic time window was closed by then. CONCLUSION Overall, data suggest that non-invasive neuromodulation by tDCS impacts neurogenesis and microglial activation as critical cellular processes influencing functional recovery during the early phase of regeneration from focal cerebral ischemia.
Collapse
Affiliation(s)
- Helene Luise Walter
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Anton Pikhovych
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Heike Endepols
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Steffen Rotthues
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Johannes Bärmann
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Heiko Backes
- Multimodal Imaging Group, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Mathias Hoehn
- Cognitive Neuroscience (INM-3), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Dirk Wiedermann
- Multimodal Imaging Group, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Bernd Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Gereon Rudolf Fink
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Cognitive Neuroscience (INM-3), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Maria Adele Rüger
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Cognitive Neuroscience (INM-3), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Michael Schroeter
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Cognitive Neuroscience (INM-3), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Jülich, Germany
| |
Collapse
|
31
|
Hirohata T, Kitano T, Saeki C, Baba K, Yoshida F, Kurihara T, Harada K, Saito S, Mochizuki H, Shimodozono M. Quantitative behavioral evaluation of a non-human primate stroke model using a new monitoring system. Front Neurosci 2022; 16:964928. [PMID: 36117634 PMCID: PMC9475201 DOI: 10.3389/fnins.2022.964928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Background Recently, the common marmoset (Callithrix jacchus) has attracted significant interest as a non-human primate stroke model. Functional impairment in non-human primate stroke models should be evaluated quantitatively and successively after stroke, but conventional observational assessments of behavior cannot fully fit this purpose. In this paper, we report a behavioral analysis using MarmoDetector, a three-dimensional motion analysis, in an ischemic stroke model using photosensitive dye, along with an observational behavioral assessment and imaging examination. Methods Ischemic stroke was induced in the left hemisphere of three marmosets. Cerebral infarction was induced by intravenous injection of rose bengal and irradiation with green light. The following day, the success of the procedure was confirmed by magnetic resonance imaging (MRI). The distance traveled, speed, activity time, and jumps/climbs were observed for 28 days after stroke using MarmoDetector. We also assessed the marmosets’ specific movements and postural abnormalities using conventional neurological scores. Results Magnetic resonance imaging diffusion-weighted and T2-weighted images showed hyperintense signals, indicating cerebral infarction in all three marmosets. MarmoDetector data showed that the both indices immediately after stroke onset and gradually improved over weeks. Neurological scores were the worst immediately after stroke and did not recover to pre-infarction levels during the observation period (28 days). A significant correlation was observed between MarmoDetector data and conventional neurological scores. Conclusion In this study, we showed that MarmoDetector can quantitatively evaluate behavioral changes in the acute to subacute phases stroke models. This technique can be practical for research on the pathophysiology of ischemic stroke and for the development of new therapeutic methods.
Collapse
Affiliation(s)
- Toshikazu Hirohata
- Department of Rehabilitation and Physical Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takaya Kitano
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Chizu Saeki
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kousuke Baba
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
- Academic Research Division, Department of Neurology, Faculty of Medicine, University of Toyama, Toyama, Japan
- *Correspondence: Kousuke Baba,
| | - Fumiaki Yoshida
- Department of Anatomy and Physiology, Faculty of Medicine, Saga University, Saga, Japan
- Fumiaki Yoshida,
| | - Takashi Kurihara
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Takashi Kurihara,
| | - Katsuhiro Harada
- Department of Rehabilitation and Physical Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Shigeyoshi Saito
- Division of Health Sciences, Department of Medical Physics and Engineering, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Megumi Shimodozono
- Department of Rehabilitation and Physical Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
32
|
Seong D, Yi S, Han S, Lee J, Park S, Hwang YH, Kim J, Kim HK, Jeon M. Target ischemic stroke model creation method using photoacoustic microscopy with simultaneous vessel monitoring and dynamic photothrombosis induction. PHOTOACOUSTICS 2022; 27:100376. [PMID: 35734368 PMCID: PMC9207728 DOI: 10.1016/j.pacs.2022.100376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/02/2022] [Indexed: 06/02/2023]
Abstract
The ischemic stroke animal model evaluates the efficacy of reperfusion and neuroprotective strategies for ischemic injuries. Various conventional methods have been reported to induce the ischemic models; however, controlling specific neurological deficits, mortality rates, and the extent of the infarction is difficult as the size of the affected region is not precisely controlled. In this paper, we report a single laser-based localized target ischemic stroke model development method by simultaneous vessel monitoring and photothrombosis induction using photoacoustic microscopy (PAM), which has minimized the infarct size at precise location with high reproducibility. The proposed method has significantly reduced the infarcted region by illuminating the precise localization. The reproducibility and validity of suggested method have been demonstrated through repeated experiments and histological analyses. These results demonstrate that our method can provide the ischemic stroke model closest to the clinical pathology for brain ischemia research from inducement, occurrence mechanisms to the recovery process.
Collapse
Affiliation(s)
- Daewoon Seong
- School of Electronic and Electrical Engineering, College of IT Engineering, Kyungpook National University, Daegu 41566, the Republic of Korea
| | - Soojin Yi
- Bio-Medical Institute, Kyungpook National University Hospital, Daegu 41404, the Republic of Korea
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Daegu 41944, the Republic of Korea
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu 41944, the Republic of Korea
| | - Sangyeob Han
- School of Electronic and Electrical Engineering, College of IT Engineering, Kyungpook National University, Daegu 41566, the Republic of Korea
- Institute of Biomedical Engineering, School of Medicine, Kyungpook National University, Daegu 41566, the Republic of Korea
| | - Jaeyul Lee
- School of Electronic and Electrical Engineering, College of IT Engineering, Kyungpook National University, Daegu 41566, the Republic of Korea
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Sungjo Park
- Pohang Innotown Center, Pohang University of Science and Technology, Pohang 37673, the Republic of Korea
| | - Yang-Ha Hwang
- Department of Neurology, School of Medicine, Kyungpook National University, Daegu 41944, the Republic of Korea
| | - Jeehyun Kim
- School of Electronic and Electrical Engineering, College of IT Engineering, Kyungpook National University, Daegu 41566, the Republic of Korea
| | - Hong Kyun Kim
- Bio-Medical Institute, Kyungpook National University Hospital, Daegu 41404, the Republic of Korea
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Daegu 41944, the Republic of Korea
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu 41944, the Republic of Korea
| | - Mansik Jeon
- School of Electronic and Electrical Engineering, College of IT Engineering, Kyungpook National University, Daegu 41566, the Republic of Korea
| |
Collapse
|
33
|
Characterization of Astrocytes in the Minocycline-Administered Mouse Photothrombotic Ischemic Stroke Model. Neurochem Res 2022; 47:2839-2855. [PMID: 35907114 DOI: 10.1007/s11064-022-03703-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/10/2022] [Accepted: 07/16/2022] [Indexed: 10/16/2022]
Abstract
Astrocytes, together with microglia, play important roles in the non-infectious inflammation and scar formation at the brain infarct during ischemic stroke. After ischemia occurs, these become highly reactive, accumulate at the infarction, and release various inflammatory signaling molecules. The regulation of astrocyte reactivity and function surrounding the infarction largely depends on intercellular communication with microglia. However, the mechanisms involved remain unclear. Furthermore, recent molecular biological studies have revealed that astrocytes are highly divergent under both resting and reactive states, whereas it has not been well reported how the communication between microglia and astrocytes affects astrocyte divergency during ischemic stroke. Minocycline, an antibiotic that reduces microglial activity, has been used to examine the functional roles of microglia in mice. In this study, we used a mouse photothrombotic ischemic stroke model to examine the characteristics of astrocytes after the administration of minocycline during ischemic stroke. Minocycline increased astrocyte reactivity and affected the localization of astrocytes in the penumbra region. Molecular characterization revealed that the induced expression of mRNA encoding the fatty acid binding protein 7 (FABP7) by photothrombosis was enhanced by the minocycline administration. Meanwhile, minocycline did not significantly affect the phenotype or class of astrocytes. The expression of Fabp7 mRNA was well correlated with that of tumor-necrosis factor α (TNFα)-encoding Tnf mRNA, indicating that a correlated expression of FABP7 from astrocytes and TNFα is suppressed by microglial activity.
Collapse
|
34
|
Poole J, Jasbi P, Pascual AS, North S, Kwatra N, Weissig V, Gu H, Bottiglieri T, Jadavji NM. Ischemic Stroke and Dietary Vitamin B12 Deficiency in Old-Aged Females: Impaired Motor Function, Increased Ischemic Damage Size, and Changed Metabolite Profiles in Brain and Cecum Tissue. Nutrients 2022; 14:2960. [PMID: 35889916 PMCID: PMC9318046 DOI: 10.3390/nu14142960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 02/04/2023] Open
Abstract
A vitamin B12 deficiency (vit. B12 def.) is common in the elderly, because of changes in metabolism. Clinical studies have reported that a vit. B12 def. results in worse outcome after stroke, and the mechanisms through which a vit. B12 def. changes the brain requires further investigation. This study investigated the role of vit. B12 def. on stroke outcome and mechanisms using aged female mice. Eighteen-month-old females were put on a control or vit. B12 def. diet for 4 weeks, after which an ischemic stroke was induced in the sensorimotor cortex. After damage, motor function was measured, the animals were euthanized, and tissues were collected for analysis. Vit. B12 def. animals had increased levels of total homocysteine in plasma and liver, and choline levels were also increased in the liver. Vit. B12 def. animals had larger damage volume in brain tissue and more apoptosis. The cecum tissue pathway analysis showed dysfunction in B12 transport. The analysis of mitochondrial metabolomics in brain tissue showed reduced levels of metabolites involved in the TCA cycle in vit. B12 def. animals. Motor function after stroke was impaired in vit. B12 def. animals. A dietary vit. B12 def. impairs motor function through increased apoptosis and changes in mitochondrial metabolism in brain tissue.
Collapse
Affiliation(s)
- Joshua Poole
- College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA; (J.P.); (S.N.)
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA; (A.S.P.); (N.K.); (V.W.)
| | - Paniz Jasbi
- College of Health Solutions, Arizona State University, Phoenix, AZ 85281, USA; (P.J.); (H.G.)
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85308, USA
| | - Agnes S. Pascual
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA; (A.S.P.); (N.K.); (V.W.)
| | - Sean North
- College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA; (J.P.); (S.N.)
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA; (A.S.P.); (N.K.); (V.W.)
| | - Neha Kwatra
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA; (A.S.P.); (N.K.); (V.W.)
- College of Dental Medicine Arizona, Midwestern University, Glendale, AZ 85308, USA
| | - Volkmar Weissig
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA; (A.S.P.); (N.K.); (V.W.)
- Department of Pharmaceutical Sciences, College of Graduate Students, Midwestern University, Glendale, AZ 85308, USA
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ 85281, USA; (P.J.); (H.G.)
- Department of Environmental Health Sciences, The Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
- Center for Translational Science, Cellular Biology and Pharmacology Department, The Herbert Wertheim College of Medicine, Florida International University, Port St. Lucie, FL 33199, USA
| | - Teodoro Bottiglieri
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott & White Research Institute, Dallas, TX 75204, USA;
| | - Nafisa M. Jadavji
- College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA; (J.P.); (S.N.)
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA; (A.S.P.); (N.K.); (V.W.)
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|
35
|
Jeon J, Kim SH, Kong E, Kim SJ, Yang JM, Lee JY, Lee J, Kim YM, Kim P. Establishment of the reproducible branch retinal artery occlusion mouse model and intravital longitudinal imaging of the retinal CX3CR1-GFP+ cells after spontaneous arterial recanalization. Front Med (Lausanne) 2022; 9:897800. [PMID: 35911406 PMCID: PMC9334526 DOI: 10.3389/fmed.2022.897800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/29/2022] [Indexed: 11/23/2022] Open
Abstract
Animal models of retinal artery occlusion (RAO) have been widely used in many studies. However, most of these studies prefer using a central retinal artery occlusion (CRAO) which is a typical global ischemia model of the retina, due to the technical limitation of producing single vessel targeted modeling with real-time imaging. A focal ischemia model, such as branch retinal artery occlusion (BRAO), is also needed for explaining interactions, including the immunological reaction between the ischemic retina and adjacent healthy retina. Accordingly, a relevant model for clinical RAO patients has been demanded to understand the pathophysiology of the RAO disease. Herein, we establish a convenient BRAO mouse model to research the focal reaction of the retina. As a photo-thrombotic agent, Rose bengal was intravenously injected into 7 week-old transgenic mice (CX3CR1-GFP) for making embolism occlusion, which causes pathology similarly to clinical cases. In an optimized condition, a 561 nm laser (13.1 mw) was projected to a targeted vessel to induce photo-thrombosis for 27 s by custom-built retinal confocal microscopy. Compared to previous BRAO models, the procedures of thrombosis generation were naturally and minimal invasively generated with real-time retinal imaging. In addition, by utilizing the self-remission characteristics of Rose bengal thrombus, a reflow of the BRAO with immunological reactions of the CX3CR1-GFP+ inflammatory cells such as the retinal microglia and monocytes was monitored and analyzed. In this models, reperfusion began on day 3 after modeling. Simultaneously, the activation of CX3CR1-GFP+ inflammatory cells, including the increase of activation marker and morphologic change, was confirmed by immunohistochemical (IHC) staining and quantitative real-time PCR. CD86 and Nox2 were prominently expressed on day 3 after the modeling. At day 7, blood flow was almost restored in the large vessels. CX3CR1-GFP+ populations in both superficial and deep layers of the retina also increased around even in the BRAO peri-ischemic area. In summary, this study successfully establishes a reproducible BRAO modeling method with convenient capabilities of easily controllable time points and selection of a specific single vessel. It can be a useful tool to analyze the behavior of inflammatory cell after spontaneous arterial recanalization in BRAO and further investigate the pathophysiology of BRAO.
Collapse
Affiliation(s)
- Jehwi Jeon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Sang-Hoon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Eunji Kong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Soo Jin Kim
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jee Myung Yang
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Dongguk University Ilsan Hospital, Ilsan, South Korea
| | - Joo Yong Lee
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Junyeop Lee
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - You-Me Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Pilhan Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- *Correspondence: Pilhan Kim,
| |
Collapse
|
36
|
Zhou J, Khateeb K, Gala A, Rahimi M, Griggs DJ, Ip Z, Yazdan-Shahmorad A. Neuroprotective Effects of Electrical Stimulation Following Ischemic Stroke in Non-Human Primates. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2022; 2022:3085-3088. [PMID: 36085944 PMCID: PMC10259874 DOI: 10.1109/embc48229.2022.9871335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Brain stimulation has emerged as a novel therapy for ischemic stroke, a major cause of brain injury that often results in lifelong disability. Although past works in rodents have demonstrated protective effects of stimulation following stroke, few of these results have been replicated in humans due to the anatomical differences between rodent and human brains and a limited understanding of stimulation-induced network changes. Therefore, we combined electrophysiology and histology to study the neuroprotective mechanisms of electrical stimulation following cortical ischemic stroke in non-human primates. To produce controlled focal lesions, we used the photothrombotic method to induce targeted vasculature damage in the sensorimotor cortices of two macaques while collecting electrocorticography (ECoG) signals bilaterally. In another two monkeys, we followed the same lesioning procedures and applied repeated electrical stimulation via an ECoG electrode adjacent to the lesion. We studied the protective effects of stimulation on neural dynamics using ECoG signal power and coherence. In addition, we performed histological analysis to evaluate the differences in lesion volume. In comparison to controls, the ECoG signals showed decreased gamma power across the sensorimotor cortices in stimulated animals. Meanwhile, Nissl staining revealed smaller lesion volumes for the stimulated group, suggesting that electrical stimulation may exert neuroprotection by suppressing post-ischemic neural activity. With the similarity between NHP and human brains, this study paves the path for developing effective stimulation-based therapy for acute stroke in clinical studies.
Collapse
|
37
|
Macrophage migration inhibitory factor (MIF) acetylation protects neurons from ischemic injury. Cell Death Dis 2022; 13:466. [PMID: 35585040 PMCID: PMC9117661 DOI: 10.1038/s41419-022-04918-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 12/14/2022]
Abstract
Ischemia-induced neuronal death leads to serious lifelong neurological deficits in ischemic stroke patients. Histone deacetylase 6 (HDAC6) is a promising target for neuroprotection in many neurological disorders, including ischemic stroke. However, the mechanism by which HDAC6 inhibition protects neurons after ischemic stroke remains unclear. Here, we discovered that genetic ablation or pharmacological inhibition of HDAC6 reduced brain injury after ischemic stroke by increasing macrophage migration inhibitory factor (MIF) acetylation. Mass spectrum analysis and biochemical results revealed that HDAC6 inhibitor or aspirin treatment promoted MIF acetylation on the K78 residue. MIF K78 acetylation suppressed the interaction between MIF and AIF, which impaired MIF translocation to the nucleus in ischemic cortical neurons. Moreover, neuronal DNA fragmentation and neuronal death were impaired in the cortex after ischemia in MIF K78Q mutant mice. Our results indicate that the neuroprotective effect of HDAC6 inhibition and aspirin treatment results from MIF K78 acetylation; thus, MIF K78 acetylation may be a therapeutic target for ischemic stroke and other neurological diseases.
Collapse
|
38
|
Zhou MY, Zhang YJ, Ding HM, Wu WF, Cai WW, Wang YQ, Geng DQ. Diprotin A TFA Exerts Neurovascular Protection in Ischemic Cerebral Stroke. Front Neurosci 2022; 16:861059. [PMID: 35615279 PMCID: PMC9125038 DOI: 10.3389/fnins.2022.861059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundIt has been established that the dipeptidyl peptidase-4 (DPP-4) inhibitor Diprotin A TFA can reduce vascular endothelial (VE)-cadherin disruption by inhibiting the increase in cleaved β-catenin in response to hypoxia, thereby protecting the vascular barrier of human umbilical vein endothelial cells. In this study, we sought to investigate the possible effect of Diprotin A TFA on the VE barrier after cerebral ischemic stroke in mice.MethodsC57BL/6J mice were divided into five groups, namely, (1) sham, (2) stroke, (3) stroke + dimethyl sulfoxide (DMSO), (4) stroke + Diprotin A TFA, and (5) stroke + Diprotin A TFA + XAV-939. First, the cerebral ischemia model was established by photothrombotic ischemia, followed by intraperitoneal injection with Diprotin A TFA and XAV-939 at doses of 70 μg/kg and 40 mg/kg 30 min once in the morning and once in the evening for 3 days. Immunofluorescence staining and Western blot methods were used to analyze the expression of vascular and blood-brain barrier (BBB)-associated molecular markers in the peri-infarct area.ResultsCompared with the vehicle control group, we found that mice injected with Diprotin A TFA exhibited reduced cerebral infarction volume, increased vascular area and length around the brain injury, increased pericyte and basement membrane coverage, upregulated expression of BBB tight junction proteins, and improved their BBB permeability, whereas the group injected with both drug and inhibitor exhibited significantly aggravated vascular injury and BBB permeability.ConclusionDiprotin A TFA can reduce VE-cadherin disruption by inhibiting ischemia-hypoxia-induced β-catenin cleavage to protect blood vessels.
Collapse
Affiliation(s)
- Ming-Yue Zhou
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ya-Jie Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hong-Mei Ding
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Neurology, Nanjing Medical University, Nanjing, China
| | - Wei-Feng Wu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wei-Wei Cai
- Department of Neurology, The Third Hospital of Huai'an, Huai'an, China
| | - Yan-Qiang Wang
- Department of Neurology, The Affiliated Hospital of Weifang Medical University, Weifang, China
- *Correspondence: Yan-Qiang Wang
| | - De-Qin Geng
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Neurology, Nanjing Medical University, Nanjing, China
- De-Qin Geng
| |
Collapse
|
39
|
Optimising the photothrombotic model of stroke in the C57BI/6 and FVB/N strains of mouse. Sci Rep 2022; 12:7598. [PMID: 35534531 PMCID: PMC9085761 DOI: 10.1038/s41598-022-11793-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
The photothrombotic stroke model relies on the interaction between photosensitive-dye and light for clot formation. Interestingly, the relationship between the length of light exposure and stroke-outcome has never been examined. This model has yet to be established in the FVB/N strain, even though stroke-outcomes are strain-specific. Therefore, this study aimed to examine the effect of different lengths of light exposure in two strains of mice on photothrombotic stroke. Male FVB/N and C57Bl/6 mice were subjected to stroke using 15, 18, or 20-min light exposure. Mice underwent functional testing for up to 7 days. Infarct volume was assessed with thionin staining, and cellular responses to injury analysed via immunofluorescence at 7-days post-stroke. Blood brain barrier (BBB) breakdown was assessed using Evans blue dye at 4.5-h post-stroke. Increasing light exposure from 15 to 20-min increased infarct volume but not functional deficit. Interestingly, there were strain-specific differences in functional outcomes, with FVB/N mice having less deficit on the hanging wire test than C57BI/6 after 15-min of light exposure. The opposite was seen in the adhesive removal test. There was no difference in the number of neurons, astrocytes, microglia, macrophages, and T cells between the strains, despite FVB/N mice demonstrating greater BBB breakdown and an enlarged spleen post-stroke. Increasing light exposure systematically increases infarct volume but does not worsen functional outcomes. FVB/N and C57Bl/6 mice exhibit subtle differences in functional outcomes post stroke, which highlights the need to choose tests which are appropriate for the mouse strain being used.
Collapse
|
40
|
Platelet-targeted thrombolysis for treatment of acute ischemic stroke. Blood Adv 2022; 7:561-574. [PMID: 35482909 PMCID: PMC9984306 DOI: 10.1182/bloodadvances.2021006691] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/28/2022] [Accepted: 04/09/2022] [Indexed: 11/20/2022] Open
Abstract
Thrombolysis with tissue-type plasminogen activator (tPA) remains the main treatment for acute ischemic stroke. Nevertheless, tPA intervention is limited by a short therapeutic window, low recanalization rates, and a risk of intracranial hemorrhage (ICH), highlighting the clinical demand for improved thrombolytic drugs. We examined a novel thrombolytic agent termed "SCE5-scuPA," comprising a single-chain urokinase plasminogen activator (scuPA) fused with a single-chain antibody (SCE5) that targets the activated glycoprotein IIb/IIIa platelet receptor, for its effects in experimental stroke. SCE5-scuPA was first tested in a whole blood clot degradation assay to show the benefit of platelet-targeted thrombolysis. The tail bleeding time, blood clearance, and biodistribution were then determined to inform the use of SCE5-scuPA in mouse models of photothrombotic stroke and middle cerebral artery occlusion against tenecteplase. The impacts of SCE5-scuPA on motor function, ICH, blood-brain barrier (BBB) integrity, and immunosuppression were evaluated. Infarct size was measured by computed tomography imaging and magnetic resonance imaging. SCE5-scuPA enhanced clot degradation ex vivo compared with its nonplatelet-targeting control. The maximal SCE5-scuPA dose that maintained hemostasis and a rapid blood clearance was determined. SCE5-scuPA administration both before and 2 hours after photothrombotic stroke reduced the infarct volume. SCE5-scuPA also improved neurologic deficit, decreased intracerebral blood deposits, preserved the BBB, and alleviated immunosuppression poststroke. In middle cerebral artery occlusion, SCE5-scuPA did not worsen stroke outcomes or cause ICH, and it protected the BBB. Our findings support the ongoing development of platelet-targeted thrombolysis with SCE5-scuPA as a novel emergency treatment for acute ischemic stroke with a promising safety profile.
Collapse
|
41
|
Seyedaghamiri F, Hosseini L, Kazmi S, Mahmoudi J, Shanehbandi D, Ebrahimi-Kalan A, Rahbarghazi R, Sadigh-Eteghad S, Farhoudi M. Varenicline improves cognitive impairment in a mouse model of mPFC ischemia: The possible roles of inflammation, apoptosis, and synaptic factors. Brain Res Bull 2022; 181:36-45. [DOI: 10.1016/j.brainresbull.2022.01.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/15/2022] [Accepted: 01/19/2022] [Indexed: 12/16/2022]
|
42
|
Kauer SD, Fink KL, Li EHF, Evans BP, Golan N, Cafferty WBJ. Inositol Polyphosphate-5-Phosphatase K ( Inpp5k) Enhances Sprouting of Corticospinal Tract Axons after CNS Trauma. J Neurosci 2022; 42:2190-2204. [PMID: 35135857 PMCID: PMC8936595 DOI: 10.1523/jneurosci.0897-21.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 11/21/2022] Open
Abstract
Failure of CNS neurons to mount a significant growth response after trauma contributes to chronic functional deficits after spinal cord injury. Activator and repressor screening of embryonic cortical neurons and retinal ganglion cells in vitro and transcriptional profiling of developing CNS neurons harvested in vivo have identified several candidates that stimulate robust axon growth in vitro and in vivo Building on these studies, we sought to identify novel axon growth activators induced in the complex adult CNS environment in vivo We transcriptionally profiled intact sprouting adult corticospinal neurons (CSNs) after contralateral pyramidotomy (PyX) in nogo receptor-1 knock-out mice and found that intact CSNs were enriched in genes in the 3-phosphoinositide degradation pathway, including six 5-phosphatases. We explored whether inositol polyphosphate-5-phosphatase K (Inpp5k) could enhance corticospinal tract (CST) axon growth in preclinical models of acute and chronic CNS trauma. Overexpression of Inpp5k in intact adult CSNs in male and female mice enhanced the sprouting of intact CST terminals after PyX and cortical stroke and sprouting of CST axons after acute and chronic severe thoracic spinal contusion. We show that Inpp5k stimulates axon growth in part by elevating the density of active cofilin in labile growth cones, thus stimulating actin polymerization and enhancing microtubule protrusion into distal filopodia. We identify Inpp5k as a novel CST growth activator capable of driving compensatory axon growth in multiple complex CNS injury environments and underscores the veracity of using in vivo transcriptional screening to identify the next generation of cell-autonomous factors capable of repairing the damaged CNS.SIGNIFICANCE STATEMENT Neurologic recovery is limited after spinal cord injury as CNS neurons are incapable of self-repair post-trauma. In vitro screening strategies exploit the intrinsically high growth capacity of embryonic CNS neurons to identify novel axon growth activators. While promising candidates have been shown to stimulate axon growth in vivo, concomitant functional recovery remains incomplete. We identified Inpp5k as a novel axon growth activator using transcriptional profiling of intact adult corticospinal tract (CST) neurons that had initiated a growth response after pyramidotomy in plasticity sensitized nogo receptor-1-null mice. Here, we show that Inpp5k overexpression can stimulate CST axon growth after pyramidotomy, stroke, and acute and chronic contusion injuries. These data support in vivo screening approaches to identify novel axon growth activators.
Collapse
Affiliation(s)
- Sierra D Kauer
- Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Kathryn L Fink
- Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Elizabeth H F Li
- Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Brian P Evans
- Regeneron Pharmaceuticals, Tarrytown, New York 10591
| | - Noa Golan
- Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520
| | - William B J Cafferty
- Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
43
|
Astrocyte-secreted chordin-like 1 regulates spine density after ischemic injury. Sci Rep 2022; 12:4176. [PMID: 35264691 PMCID: PMC8907233 DOI: 10.1038/s41598-022-08031-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 03/01/2022] [Indexed: 11/09/2022] Open
Abstract
Ischemic injury occurs when the brain is deprived of blood flow, preventing cells from receiving essential nutrients. The injury core is the brain region directly deprived and is surrounded by the peri-infarct area, the region with recovery potential. In the peri-infarct area neurons undergo acute loss of dendritic spines, which modifies synaptic plasticity and determines neuronal survival. Astrocytes can be protective or detrimental to the ischemic injury response depending on the specific stage, yet we lack clear understanding of the underlying mechanisms. Chordin-like 1 (Chrdl1) is an astrocyte-secreted protein that promotes synaptic maturation and limits experience-dependent plasticity in the mouse visual cortex. Given this plasticity-limiting function we asked if Chrdl1 regulates the response to ischemic injury, modelled using photothrombosis (PT). We find that Chrdl1 mRNA is upregulated in astrocytes in the peri-infarct area in both acute and sub-acute phases post-PT. To determine the impact of increased Chrdl1 on the response to PT we analyzed Chrdl1 knock-out mice. We find that absence of Chrdl1 prevents ischemia-induced spine loss in the peri-infarct area and reduces cell death in the core, without impacting gliosis. These findings highlight the important role of astrocyte-secreted proteins in regulating structural plasticity in response to brain ischemic injuries.
Collapse
|
44
|
Li N, Zhang X, Zhai J, Yin J, Ma K, Wang R, Qin X, Li Y, Dong X, Wang S. Isoflurane and Netrin-1 combination therapy enhances angiogenesis and neurological recovery by improving the expression of HIF-1α-Netrin-1-UNC5B/VEGF cascade to attenuate cerebral ischemia injury. Exp Neurol 2022; 352:114028. [PMID: 35247371 DOI: 10.1016/j.expneurol.2022.114028] [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: 01/22/2022] [Revised: 02/19/2022] [Accepted: 02/26/2022] [Indexed: 12/01/2022]
Abstract
Ischemic stroke (IS) causes many morbidities and deaths worldwide. However, the current monotherapy strategy is not satisfactory. Therefore, it is urgent to explore possible combined treatment methods. Although both isoflurane (ISO) and Netrin-1 (NT-1) have angiogenesis and neuroprotective effects, it is still unclear whether combining ISO with NT-1 will provide a positive effect and the possible mechanism of action. In this study, we used a photochemical (PTI) method to establish a mouse ischemic stroke model. ISO and NT-1 were used to treat the mice for 1 week. The adhesive removal test, Morris water maze test, modified neurological severity scores and triphenyl tetrazolium chloride staining were performed to test the treatment effect. Western blotting was performed to assess protein expression, immunofluorescence staining (IF) and immunohistochemical staining (IHC) was used to evaluate angiogenesis. The results suggested that combining ISO with NT-1 resulted in a better therapeutic effect than ISO or NT-1 treatment after PTI injury (all P < 0.01). The protein expression of VEGFA and CD34 in the ISO + NT-1 group was significantly increased compared with that in the other groups (all P < 0.05). IF and IHC also showed that the ISO + NT-1 group significantly improved angiogenesis (all P < 0.01). YC-1 (an HIF-1α inhibitor) and Unc5B siRNA were used to inhibit the expression of HIF-1α and UNC5B before and after combination ISO and NT-1 treatment. The combined inhibition group not only expressed the least VEGFA and CD34 but also expressed the least HIF-1α, UNC5B, FAK, and β-catenin in all groups (all P < 0.05). Most importantly, angiogenesis and neurological recovery were also significantly decreased by inhibiting HIF-1α and UNC5B (all P < 0.05). In conclusion, our results suggested that ISO combined with NT-1 could promote angiogenesis, recover long-term neurobehavioral function, and attenuate cerebral ischemia injury by activating the HIF-1α-Netrin-1-UNC5B/VEGF cascade.
Collapse
Affiliation(s)
- Nian Li
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Xu Zhang
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Jingwen Zhai
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Jiangwen Yin
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Ketao Ma
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, China; Department of Physiology, School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, China
| | - Ruixue Wang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, China; Department of Physiology, School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, China
| | - Xinlei Qin
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, China; Department of Physiology, School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, China
| | - Yan Li
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, China; Department of Physiology, School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, China
| | - Xiwei Dong
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China.
| | - Sheng Wang
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
45
|
Optical Modalities for Research, Diagnosis, and Treatment of Stroke and the Consequent Brain Injuries. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12041891] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Stroke is the second most common cause of death and third most common cause of disability worldwide. Therefore, it is an important disease from a medical standpoint. For this reason, various studies have developed diagnostic and therapeutic techniques for stroke. Among them, developments and applications of optical modalities are being extensively studied. In this article, we explored three important optical modalities for research, diagnostic, and therapeutics for stroke and the brain injuries related to it: (1) photochemical thrombosis to investigate stroke animal models; (2) optical imaging techniques for in vivo preclinical studies on stroke; and (3) optical neurostimulation based therapy for stroke. We believe that an exploration and an analysis of previous studies will help us proceed from research to clinical applications of optical modalities for research, diagnosis, and treatment of stroke.
Collapse
|
46
|
Nucci MP, Oliveira FA, Ferreira JM, Pinto YO, Alves AH, Mamani JB, Nucci LP, Valle NME, Gamarra LF. Effect of Cell Therapy and Exercise Training in a Stroke Model, Considering the Cell Track by Molecular Image and Behavioral Analysis. Cells 2022; 11:cells11030485. [PMID: 35159294 PMCID: PMC8834410 DOI: 10.3390/cells11030485] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/12/2022] Open
Abstract
The goal of this study is to see how combining physical activity with cell treatment impacts functional recovery in a stroke model. Molecular imaging and multimodal nanoparticles assisted in cell tracking and longitudinal monitoring (MNP). The viability of mesenchymal stem cell (MSC) was determined using a 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay and bioluminescent image (BLI) after lentiviral transduction and MNP labeling. At random, the animals were divided into 5 groups (control-G1, and experimental G2-G5). The photothrombotic stroke induction was confirmed by local blood perfusion reduction and Triphenyltetrazolium chloride (TTC), and MSC in the G3 and G5 groups were implanted after 24 h, with BLI and near-infrared fluorescence image (NIRF) tracking these cells at 28 h, 2, 7, 14, and 28 days. During a 28-day period, the G5 also conducted physical training, whereas the G4 simply did the training. At 0, 7, 14, and 28 days, the animals were functionally tested using a cylinder test and a spontaneous motor activity test. MNP internalization in MSC was confirmed using brightfield and fluorescence microscopy. In relation to G1 group, only 3% of cell viability reduced. The G2–G5 groups showed more than 69% of blood perfusion reduction. The G5 group performed better over time, with a progressive recovery of symmetry and an increase of fast vertical movements. Up to 7 days, BLI and NIRF followed MSC at the damaged site, demonstrating a signal rise that could be connected to cell proliferation at the injury site during the acute phase of stroke. Local MSC therapy mixed with physical activity resulted in better results in alleviating motor dysfunction, particularly during the acute period. When it comes to neurorehabilitation, this alternative therapy could be a suitable fit.
Collapse
Affiliation(s)
- Mariana P. Nucci
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil; (M.P.N.); (F.A.O.); (J.M.F.); (Y.O.P.); (A.H.A.); (J.B.M.); (N.M.E.V.)
- LIM44, Hospital das Clínicas da Faculdade Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - Fernando A. Oliveira
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil; (M.P.N.); (F.A.O.); (J.M.F.); (Y.O.P.); (A.H.A.); (J.B.M.); (N.M.E.V.)
| | - João M. Ferreira
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil; (M.P.N.); (F.A.O.); (J.M.F.); (Y.O.P.); (A.H.A.); (J.B.M.); (N.M.E.V.)
| | - Yolanda O. Pinto
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil; (M.P.N.); (F.A.O.); (J.M.F.); (Y.O.P.); (A.H.A.); (J.B.M.); (N.M.E.V.)
| | - Arielly H. Alves
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil; (M.P.N.); (F.A.O.); (J.M.F.); (Y.O.P.); (A.H.A.); (J.B.M.); (N.M.E.V.)
| | - Javier B. Mamani
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil; (M.P.N.); (F.A.O.); (J.M.F.); (Y.O.P.); (A.H.A.); (J.B.M.); (N.M.E.V.)
| | - Leopoldo P. Nucci
- Centro Universitário do Planalto Central, Brasília 72445-020, Brazil;
| | - Nicole M. E. Valle
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil; (M.P.N.); (F.A.O.); (J.M.F.); (Y.O.P.); (A.H.A.); (J.B.M.); (N.M.E.V.)
| | - Lionel F. Gamarra
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil; (M.P.N.); (F.A.O.); (J.M.F.); (Y.O.P.); (A.H.A.); (J.B.M.); (N.M.E.V.)
- Correspondence: ; Tel.: +55-11-2151-0243
| |
Collapse
|
47
|
Kang J, Liu X, Cao S, Zeiler SR, Graham EM, Boctor EM, Koehler RC. Transcranial photoacoustic characterization of neurovascular physiology during early-stage photothrombotic stroke in neonatal piglets in vivo. J Neural Eng 2022; 18:10.1088/1741-2552/ac4596. [PMID: 34937013 PMCID: PMC9112348 DOI: 10.1088/1741-2552/ac4596] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 12/22/2021] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Perinatal ischemic stroke is estimated to occur in 1/2300-1/5000 live births, but early differential diagnosis from global hypoxia-ischemia is often difficult. In this study, we tested the ability of a hand-held transcranial photoacoustic (PA) imaging probe to non-invasively detect a focal photothrombotic stroke (PTS) within 2 h of stroke onset in a gyrencephalic piglet brain. APPROACH About 17 stroke lesions of approximately 1 cm2area were introduced randomly in anterior or posterior cortex via the light/dye PTS technique in anesthetized neonatal piglets (n= 11). The contralateral non-ischemic region served as control tissue for discrimination contrast for the PA hemoglobin metrics: oxygen saturation, total hemoglobin (tHb), and individual quantities of oxygenated and deoxygenated hemoglobin (HbO2and HbR). MAIN RESULTS The PA-derived tissue oxygen saturation at 2 h yielded a significant separation between control and affected regions-of-interest (p< 0.0001), which were well matched with 24 h post-stroke cerebral infarction confirmed in the triphenyltetrazolium chloride-stained image. The quantity of HbO2also displayed a significant contrast (p= 0.021), whereas tHb and HbR did not. The analysis on receiver operating characteristic curves and multivariate data analysis also agreed with the results above. SIGNIFICANCE This study shows that a hand-held transcranial PA neuroimaging device can detect a regional thrombotic stroke in the cerebral cortex of a neonatal piglet. In particular, we conclude that the oxygen saturation metric can be used alone to identify regional stroke lesions. The lack of change in tHb may be related to arbitrary hand-held imaging configuration and/or entrapment of red blood cells within the thrombotic stroke.
Collapse
Affiliation(s)
- Jeeun Kang
- Laboratory for Computational Sensing and Robotics, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, United States of America,These authors equally contributed
| | - Xiuyun Liu
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, United States of America,These authors equally contributed
| | - Suyi Cao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, United States of America
| | - Steven R Zeiler
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, United States of America
| | - Ernest M Graham
- Division of Maternal-Fetal Medicine, Department of Gynecology-Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States of America,Neuroscience Intensive Care Nursery Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States of America
| | - Emad M Boctor
- Laboratory for Computational Sensing and Robotics, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, United States of America,Authors to whom any correspondence should be addressed. and
| | - Raymond C Koehler
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, United States of America,Authors to whom any correspondence should be addressed. and
| |
Collapse
|
48
|
Cambiaghi M, Cherchi L, Comai S. Photothrombotic Mouse Models for the Study of Melatonin as a Therapeutic Tool After Ischemic Stroke. Methods Mol Biol 2022; 2550:433-441. [PMID: 36180711 DOI: 10.1007/978-1-0716-2593-4_42] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Melatonin is a potent neuroprotective agent which has shown therapeutic effects in animal models of brain injury such as stroke. Currently, there are few effective treatments for the therapeutics of stroke, the second leading cause of death and a major cause of disability worldwide. As demonstrated by the high number of publications during the last two decades, there is growing interest in understanding how and if melatonin could be a possible drug for stroke in humans, given also its very low and limited toxicity. Here, we describe the detailed protocol for performing the photothrombotic model of stroke which involves the occlusion of small cerebral vessels caused by the photoactivation of the previously injected light-sensitive dye Rose Bengal. Importantly, this model allows for the study of cellular and molecular mechanisms underlying the pathophysiology of stroke and thus can be used for investigating the neuropharmacological role of melatonin and the melatonin system in stroke. In particular, future research is warranted to demonstrate how and if melatonin impacts neurodegeneration, neuroprotection, and neuro-regeneration occurring after the brain injury caused by the occlusion of cerebral vessels.
Collapse
Affiliation(s)
- Marco Cambiaghi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Laura Cherchi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Comai
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy.
- Department of Biomedical Sciences, University of Padua, Padua, Italy.
- Department of Psychiatry, McGill University, Montreal, Canada.
| |
Collapse
|
49
|
A novel BRET based genetic coded biosensor for apoptosis detection at deep tissue level in live animal. Apoptosis 2021; 26:628-638. [PMID: 34748127 DOI: 10.1007/s10495-021-01693-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2021] [Indexed: 10/19/2022]
Abstract
ANNEXIN V belongs to a family of phospholipid binding proteins which is able to bind to negatively charged phospholipids such as phosphatidylserine (PS) in the presence of a high affinity Ca2+ ion. When apoptosis occurs, even at early stage, PS will be exposed to the outside of the cell surface from the cytoplasm side of membrane leaflets., Therefore ANNEXIN V has been suggested as a bio-marker for imaging early apoptotic events of various cell death including those in disease conditions. However, most ANNEXIN V-based apoptotic detecting techniques were in vitro approaches. Here, we presented a new BRET (Bioluminescence Resonance Energy Transfer) based genetic coded biosensor by fusing ANNEXIN V and a BRET version of NanoLuc (teLuc) for both in vitro and in vivo apoptosis detection. The BRET feature of this new sensor makes it convenient to be applied to both conventional fluorescent-based in vitro apoptosis detection and bioluminescence-based animal live imaging for in vivo study. Because of its robust bioluminescence signal, it is possible to perform the evaluation of the disease-induced apoptotic damage and recovery process directly at deep tissue level in live animal.
Collapse
|
50
|
P75 neurotrophin receptor controls subventricular zone neural stem cell migration after stroke. Cell Tissue Res 2021; 387:415-431. [PMID: 34698916 PMCID: PMC8975773 DOI: 10.1007/s00441-021-03539-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 10/04/2021] [Indexed: 12/23/2022]
Abstract
Stroke is the leading cause of adult disability. Endogenous neural stem/progenitor cells (NSPCs) originating from the subventricular zone (SVZ) contribute to the brain repair process. However, molecular mechanisms underlying CNS disease-induced SVZ NSPC-redirected migration to the lesion area are poorly understood. Here, we show that genetic depletion of the p75 neurotrophin receptor (p75NTR−/−) in mice reduced SVZ NSPC migration towards the lesion area after cortical injury and that p75NTR−/− NSPCs failed to migrate upon BDNF stimulation in vitro. Cortical injury rapidly increased p75NTR abundance in SVZ NSPCs via bone morphogenetic protein (BMP) receptor signaling. SVZ-derived p75NTR−/− NSPCs revealed an altered cytoskeletal network- and small GTPase family-related gene and protein expression. In accordance, BMP-treated non-migrating p75NTR−/− NSPCs revealed an altered morphology and α-tubulin expression compared to BMP-treated migrating wild-type NSPCs. We propose that BMP-induced p75NTR abundance in NSPCs is a regulator of SVZ NSPC migration to the lesion area via regulation of the cytoskeleton following cortical injury.
Collapse
|