1
|
Shanks MJ, Cirillo J, Stinear CM, Byblow WD. A novel TMS framework for assessing neurophysiological recovery at the subacute stage after stroke. Clin Neurophysiol 2025; 171:82-94. [PMID: 39889484 DOI: 10.1016/j.clinph.2025.01.010] [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: 05/01/2024] [Revised: 10/29/2024] [Accepted: 01/14/2025] [Indexed: 02/03/2025]
Abstract
OBJECTIVE To use peri-threshold transcranial magnetic stimulation (TMS) intensities to elicit motor evoked potentials (MEPs) during the subacute stage after stroke and assess their association with upper limb motor recovery. METHODS Twenty-five MEP+ patients participated in three sessions at 1, 3, and 6 months post-stroke. Single-pulse TMS across a range of stimulation intensities was used to elicit MEPs in four muscles of the paretic and non-paretic upper limb. At each timepoint, threshold matrices were constructed based on MEP amplitude and persistence. A matrix element was suprathreshold if five out of ten stimulations elicited MEPs ≥ 50 μV. A subthreshold element produced MEPs below this criterion. Dexterity was assessed using the nine hole peg test. RESULTS There were fewer suprathreshold, and more subthreshold elements on the paretic compared to the non-paretic side. The number of suprathreshold elements on the paretic side increased between 1 and 6 months post-stroke. Neither sub- nor supra-threshold elements were associated with dexterity recovery. CONCLUSION The proportion of sub- and supra-threshold elements reflect neurophysiological recovery during the subacute stage after stroke. A threshold matrix framework can identify patients with stable versus dynamic neurophysiology post-stroke. SIGNIFICANCE A compositional analysis framework can quantify neurophysiological recovery after stroke.
Collapse
Affiliation(s)
- Maxine J Shanks
- Department of Exercise Sciences, University of Auckland, New Zealand; Centre for Brain Research, University of Auckland, New Zealand
| | - John Cirillo
- Department of Exercise Sciences, University of Auckland, New Zealand; Discipline of Physiology, School of Biomedicine, The University of Adelaide, Australia
| | - Cathy M Stinear
- Centre for Brain Research, University of Auckland, New Zealand; Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Winston D Byblow
- Department of Exercise Sciences, University of Auckland, New Zealand; Centre for Brain Research, University of Auckland, New Zealand.
| |
Collapse
|
2
|
Raffaele S, Clausen BH, Mannella FC, Wirenfeldt M, Marangon D, Tidgen SB, Corradini S, Madsen K, Lecca D, Abbracchio MP, Lambertsen KL, Fumagalli M. Characterisation of GPR17-expressing oligodendrocyte precursors in human ischaemic lesions and correlation with reactive glial responses. J Pathol 2025; 265:226-243. [PMID: 39703181 PMCID: PMC11717493 DOI: 10.1002/path.6381] [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: 06/03/2024] [Revised: 10/14/2024] [Accepted: 11/20/2024] [Indexed: 12/21/2024]
Abstract
White matter damage and subsequent demyelination significantly contribute to long-term functional impairment after ischaemic stroke. Identifying novel pharmacological targets to restore myelin integrity by promoting the maturation of oligodendrocyte precursor cells (OPCs) into new myelinating oligodendrocytes may open new perspectives for ischaemic stroke treatment. In this respect, previous studies highlighted the role of the G protein-coupled membrane receptor 17 (GPR17) as a key regulator of OPC differentiation in experimental models of brain injury, including ischaemic stroke. To determine the translational value of GPR17 as a possible target in the context of human disease, we exploited immunohistochemistry to characterise the distribution of GPR17-expressing cells in brain tissue samples from ischaemic stroke cases and correlated it with the reactive state of neighbouring glial cells. The results showed that GPR17 specifically decorates a subpopulation of differentiation-committed OPCs, labelled by the peculiar marker breast carcinoma-amplified sequence 1 (BCAS1), that accumulates in the peri-infarct region in the later stages after the ischaemic event. Interestingly, the response of GPR17-expressing cells appears to be paralleled by the switch of reactive microglia/macrophages from a phagocytic to a dystrophic phenotype and by astrocytic scar formation. A negative correlation was found between GPR17-expressing OPCs and reactive microglia/macrophages and astrocytes surrounding chronic ischaemic lesions in female subjects, while the same relationship was less pronounced in males. These results were reinforced by bioinformatic analysis of a publicly available transcriptomic dataset, which implicated a possible role of inflammation and defective neuron-to-OPC communication in remyelination failure after ischaemic damage. Hence, these data strengthen the relevance of GPR17-based remyelinating therapies for the treatment of ischaemic stroke. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Stefano Raffaele
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’Università degli Studi di MilanoMilanItaly
| | - Bettina Hjelm Clausen
- Department of Neurobiology Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
- Department of Clinical Research, Brain Research – Inter Disciplinary Guided Excellence (BRIDGE)University of Southern DenmarkOdenseDenmark
- Odense Patient data Explorative Network (OPEN), Department of Clinical Research, Odense University HospitalUniversity of Southern DenmarkOdenseDenmark
| | - Francesca Carolina Mannella
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’Università degli Studi di MilanoMilanItaly
| | - Martin Wirenfeldt
- Department of Clinical Research, Brain Research – Inter Disciplinary Guided Excellence (BRIDGE)University of Southern DenmarkOdenseDenmark
- Odense Patient data Explorative Network (OPEN), Department of Clinical Research, Odense University HospitalUniversity of Southern DenmarkOdenseDenmark
- Department of PathologySouth Denmark University HospitalOdenseDenmark
| | - Davide Marangon
- Department of Pharmaceutical SciencesUniversità degli Studi di MilanoMilanItaly
| | - Sarah Boe Tidgen
- Department of Neurobiology Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Silvia Corradini
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’Università degli Studi di MilanoMilanItaly
- Department of Neurobiology Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Kirsten Madsen
- Department of PathologySouth Denmark University HospitalOdenseDenmark
- Department of Cardiovascular and Renal Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Davide Lecca
- Department of Pharmaceutical SciencesUniversità degli Studi di MilanoMilanItaly
| | | | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
- Department of Clinical Research, Brain Research – Inter Disciplinary Guided Excellence (BRIDGE)University of Southern DenmarkOdenseDenmark
- Odense Patient data Explorative Network (OPEN), Department of Clinical Research, Odense University HospitalUniversity of Southern DenmarkOdenseDenmark
- Department of NeurologyOdense University HospitalOdenseDenmark
| | - Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’Università degli Studi di MilanoMilanItaly
| |
Collapse
|
3
|
Lv J, Jiao Y, Zhao X, Kong X, Chen Y, Li L, Chen X, Tao X, Dong D. Examining the Impact of Microglia on Ischemic Stroke With an Emphasis on the Metabolism of Immune Cells. CNS Neurosci Ther 2025; 31:e70229. [PMID: 39945118 PMCID: PMC11822359 DOI: 10.1111/cns.70229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/13/2024] [Accepted: 01/11/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Ischemic stroke, a major cause of disability and the second leading cause of death, poses a significant public health challenge. Post-stroke inflammation can harm the blood-brain barrier and worsen neurological deficits, which are key factors in neuronal damage in patients with ischemic stroke. Microglia are crucial in the central nervous system, involved in inflammation, neuronal damage, and repair after cerebral ischemia. While cellular immune metabolism has been widely studied, its role in ischamic stroke remains unclear. AIM This review aims to examine how inflammation affects the phenotypic characteristics of immune cells after ischemic stroke and to explore the effects of the immune metabolic microenvironment on the phenotypic profiles and functions of microglia in ischemic stroke. METHOD The review refers to the available literature in PubMed, searching for critical terms related to Ischemic stroke, neuroinflammation, microglia, and immunometabolism. RESULT In this review, we found that during stroke progression, microglia can dynamically switch between pro-inflammatory and anti-inflammatory phenotypes. Microglial glycometabolism includes oxidative phosphorylation and glycolysis, and lipid metabolism involves lipid synthesis and breakdown. Modulating the production of inflammatory mediator precursors can induce an anti-inflammatory phenotype in microglia. CONCLUSION Thus, studying microglial metabolic pathways and their products may offer new insights for ischemic stroke treatment.
Collapse
Affiliation(s)
- Jing Lv
- Department of PharmacyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
- College of PharmacyDalian Medical UniversityDalianChina
| | - Yang Jiao
- Department of NeurologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
- Dalian Innovation Institute of Stem Cell and Precision MedicineDalianChina
| | - Xinya Zhao
- Department of PharmacyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
- College of PharmacyDalian Medical UniversityDalianChina
| | - Xin Kong
- Department of PharmacyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
- Dalian Innovation Institute of Stem Cell and Precision MedicineDalianChina
| | - Yanwei Chen
- Department of PharmacyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Lu Li
- Department of PharmacyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Xuyang Chen
- Department of PharmacyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Xufeng Tao
- Department of PharmacyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Deshi Dong
- Department of PharmacyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
4
|
Fu Q, Yu Q, Luo H, Liu Z, Ma X, Wang H, Cheng Z. Protective effects of wogonin in the treatment of central nervous system and degenerative diseases. Brain Res Bull 2025; 221:111202. [PMID: 39814324 DOI: 10.1016/j.brainresbull.2025.111202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/29/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025]
Abstract
Wogonin, an O-methylated flavonoid extracted from Scutellaria baicalensis, has demonstrated profound neuroprotective effects in a range of central nervous system (CNS) diseases. This review elucidates the pharmacological mechanisms underlying the protective effects of wogonin in CNS diseases, including ischemic stroke, hemorrhagic stroke, traumatic brain injury, epilepsy, anxiety, neurodegenerative diseases, and CNS infections. Wogonin modulates key signaling pathways, such as the MAPK, NF-κB, and ROS pathways, contributing to its anti-inflammatory, antioxidant, and antiapoptotic properties. In ischemic stroke models, wogonin reduces infarct size and enhances neurological outcomes by mitigating inflammation and oxidative stress. For patients with hemorrhagic stroke and traumatic brain injury, it accelerates hematoma regression, mitigates secondary brain damage, and promotes neurogenesis, making it an entirely new treatment option for patients with limited access to this type of therapy. Its anticonvulsant and anxiolytic effects are mediated through GABA-A receptor modulation. Moreover, wogonin shows promise in treating neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease by promoting autophagy and reducing neuroinflammation. Additionally, it exhibits antiviral properties, offering potential benefits against CNS infections. Despite extensive preclinical evidence, further clinical studies are warranted to confirm its efficacy and safety in humans. This review highlights the great therapeutic potential of wogonin in terms of CNS protection. However, despite the substantial preclinical evidence, further large-scale clinical studies are necessary. Future researchers need to further explore the long-term efficacy and safety of wogonin in clinical trials and translate it for early application in the clinical treatment of true CNS disorders.
Collapse
Affiliation(s)
- Qingan Fu
- Department of Neurology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, Jiangxi 330006, China; Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, Jiangxi 330006, China
| | - Qingyun Yu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, Jiangxi 330006, China
| | - Hongdan Luo
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhekang Liu
- Rheumatology and Immunology Department, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xiaowei Ma
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, Jiangxi 330006, China
| | - Huijian Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, Jiangxi 330006, China
| | - Zhijuan Cheng
- Department of Neurology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
5
|
Della-Flora Nunes G, Osso LA, Haynes JA, Conant L, Thornton MA, Stockton ME, Brassell KA, Morris A, Mancha Corchado YI, Gaynes JA, Chavez AR, Woerner MB, MacKenna DA, Alavi A, Danks A, Poleg-Polsky A, Gandhi R, Vivian JA, Denman DJ, Hughes EG. Incomplete remyelination via therapeutically enhanced oligodendrogenesis is sufficient to recover visual cortical function. Nat Commun 2025; 16:732. [PMID: 39820244 PMCID: PMC11739692 DOI: 10.1038/s41467-025-56092-6] [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: 02/20/2024] [Accepted: 01/09/2025] [Indexed: 01/19/2025] Open
Abstract
Myelin loss induces neural dysfunction and contributes to the pathophysiology of neurodegenerative diseases, injury conditions, and aging. Because remyelination is often incomplete, better understanding endogenous remyelination and developing remyelination therapies that restore neural function are clinical imperatives. Here, we use in vivo two-photon microscopy and electrophysiology to study the dynamics of endogenous and therapeutic-induced cortical remyelination and functional recovery after cuprizone-mediated demyelination in mice. We focus on the visual pathway, which is uniquely positioned to provide insights into structure-function relationships during de/remyelination. We show endogenous remyelination is driven by recent oligodendrocyte loss and is highly efficacious following mild demyelination, but fails to restore the oligodendrocyte population when high rates of oligodendrocyte loss occur quickly. Testing a thyromimetic (LL-341070) compared to clemastine, we find it better enhances oligodendrocyte gain and hastens recovery of neuronal function. The therapeutic benefit of the thyromimetic is temporally restricted, and it acts exclusively following moderate to severe demyelination, eliminating the endogenous remyelination deficit. However, we find regeneration of oligodendrocytes and myelin to healthy levels is not necessary for recovery of visual neuronal function. These findings advance our understanding of remyelination and its impact on functional recovery to inform future therapeutic strategies.
Collapse
Affiliation(s)
- Gustavo Della-Flora Nunes
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lindsay A Osso
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Johana A Haynes
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lauren Conant
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Michael A Thornton
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Michael E Stockton
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Katherine A Brassell
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Amanda Morris
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Yessenia I Mancha Corchado
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - John A Gaynes
- Department of Physiology and Biophysics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Anthony R Chavez
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | | | | | - Aryan Alavi
- Autobahn Therapeutics Inc, San Diego, CA, USA
| | - Anne Danks
- Autobahn Therapeutics Inc, San Diego, CA, USA
| | - Alon Poleg-Polsky
- Department of Physiology and Biophysics, University of Colorado School of Medicine, Aurora, CO, USA
| | | | | | - Daniel J Denman
- Department of Physiology and Biophysics, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Ethan G Hughes
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
6
|
Iadecola C, Anrather J. The immunology of stroke and dementia. Immunity 2025; 58:18-39. [PMID: 39813992 PMCID: PMC11736048 DOI: 10.1016/j.immuni.2024.12.008] [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: 09/03/2024] [Revised: 12/04/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025]
Abstract
Ischemic stroke and vascular cognitive impairment, caused by a sudden arterial occlusion or more subtle but protracted vascular insufficiency, respectively, are leading causes of morbidity and mortality worldwide with limited therapeutic options. Innate and adaptive immunity have long been implicated in neurovascular injury, but recent advances in methodology and new experimental approaches have shed new light on their contributions. A previously unappreciated dynamic interplay of brain-resident, meningeal, and systemic immune cells with the ischemic brain and its vasculature has emerged, and new insights into the frequent overlap between vascular and Alzheimer pathology have been provided. Here, we critically review these recent findings, place them in the context of current concepts on neurovascular pathologies and Alzheimer's disease, and highlight their impact on recent stroke and Alzheimer therapies.
Collapse
Affiliation(s)
- Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
7
|
Pang B, Wu L, Peng Y. In vitro modelling of the neurovascular unit for ischemic stroke research: Emphasis on human cell applications and 3D model design. Exp Neurol 2024; 381:114942. [PMID: 39222766 DOI: 10.1016/j.expneurol.2024.114942] [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: 03/15/2024] [Revised: 07/16/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Ischemic stroke has garnered global medical attention as one of the most serious cerebrovascular diseases. The mechanisms involved in both the development and recovery phases of ischemic stroke are complex, involving intricate interactions among different types of cells, each with its own unique functions. To better understand the possible pathogenesis, neurovascular unit (NVU), a concept comprising neurons, endothelial cells, mural cells, glial cells, and extracellular matrix components, has been used in analysing various brain diseases, particularly in ischemic stroke, aiming to depict the interactions between cerebral vasculature and neural cells. While in vivo models often face limitations in terms of reproducibility and the ability to precisely mimic human pathophysiology, it is now important to establish in vitro NVU models for ischemic stroke research. In order to accurately portray the pathological processes occurring within the brain, a diverse array of NVU 2D and 3D in vitro models, each possessing unique characteristics and advantages, have been meticulously developed. This review presents a comprehensive overview of recent advancements in in vitro models specifically tailored for investigating ischemic stroke. Through a systematic categorization of these developments, we elucidate the intricate links between NVU components and the pathogenesis of ischemic stroke. Furthermore, we explore the distinct advantages offered by innovative NVU models, notably 3D models, which closely emulate in vivo conditions. Additionally, an examination of current therapeutic modalities for ischemic stroke developed utilizing in vitro NVU models is provided. Serving as a valuable reference, this review aids in the design and implementation of effective in vitro models for ischemic stroke research.
Collapse
Affiliation(s)
- Bo Pang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Lei Wu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ying Peng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
8
|
Li ZR, Wang YY, Wang ZH, Qin QL, Huang C, Shi GS, He HY, Deng YH, He XY, Zhao XM. The positive role of transforming growth factor-β1 in ischemic stroke. Cell Signal 2024; 121:111301. [PMID: 39019338 DOI: 10.1016/j.cellsig.2024.111301] [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: 05/07/2024] [Revised: 07/05/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
Ischemic stroke is one of the most disabling and fatal diseases around the world. The damaged brain tissues will undergo excessive autophagy, vascular endothelial cells injury, blood-brain barrier (BBB) impairment and neuroinflammation after ischemic stroke. However, there is no unified viewpoint on the underlying mechanism of brain damage. Transforming growth factor-β1 (TGF-β1), as a multi-functional cytokine, plays a crucial role in the intricate pathological processes and helps maintain the physiological homeostasis of brain tissues through various signaling pathways after ischemic stroke. In this review, we summarize the protective role of TGF-β1 in autophagic flux, BBB, vascular remodeling, neuroinflammation and other aspects after ischemic stroke. Based on the review, we believe that TGF-β1 could serve as a key target for treating ischemic stroke.
Collapse
Affiliation(s)
- Zi-Rong Li
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Yong-Yan Wang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Zi-Han Wang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Qi-Lin Qin
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Cheng Huang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Guang-Sen Shi
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Hong-Yun He
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China; Anning First People's Hospital Affiliated to Kunming University of Science and Technology, Kunming, China.
| | - Yi-Hao Deng
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Xiu-Ying He
- Department of Anesthesiology, Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, China.
| | - Xiao-Ming Zhao
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China; Anning First People's Hospital Affiliated to Kunming University of Science and Technology, Kunming, China.
| |
Collapse
|
9
|
Xin Q, Xu F, Ma Z, Wu J. β-Caryophyllene mitigates ischemic stroke-induced white matter lesions by inhibiting pyroptosis. Exp Cell Res 2024; 442:114214. [PMID: 39159913 DOI: 10.1016/j.yexcr.2024.114214] [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: 05/14/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
β-Caryophyllene (BCP), a selective agonist for cannabinoid receptor 2 (CB2R), has demonstrated promising protective effects in various pathological conditions. However, the neuroprotective effects of BCP on white matter damage induced by ischemic stroke have not been elucidated previously. In this study, we find that BCP not only improves sensorimotor and cognitive function via CB2R but also mitigates white matter lesions in mice following ischemic stroke. Furthermore, BCP enhances the viability of MO3.13 oligodendrocytes after oxygen-glucose deprivation and reoxygenation (OGD/R), attenuating OGD/R-induced cellular damage and pyroptosis. Notably, these protective effects of BCP are partially enhanced by the NLRP3 inhibitor MCC950 and counteracted by the NLRP3 activator nigericin. In addition, nigericin significantly exacerbates neurological outcomes and increases white matter lesions following BCP treatment in middle cerebral artery occlusion (MCAO) mice. These results suggest that BCP may ameliorate neurological deficits and white matter damage induced by cerebral ischemia through inhibiting NLRP3-mediated pyroptosis.
Collapse
Affiliation(s)
- Qing Xin
- Institute of Brain Science and Diseases, And Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China; Institute of Neurobiology, Jining Medical University, Jining, 272000, China
| | - Fei Xu
- Department of Vascular Surgery, Jining NO.1 People's Hospital, Jining, 272000, China
| | - Zegang Ma
- Institute of Brain Science and Diseases, And Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Jie Wu
- Institute of Brain Science and Diseases, And Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
10
|
Khor KL, Kumarasuriar V, Tan KW, Ooi PB, Chia YC. Effects of fruit and vegetable intake on memory and attention: a systematic review of randomized controlled trials. Syst Rev 2024; 13:151. [PMID: 38849879 PMCID: PMC11157787 DOI: 10.1186/s13643-024-02547-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 04/26/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Memory and attention are important for daily functioning, and their function deteriorates due to aging. However, fruit and vegetable consumption are one of the protective factors against deterioration in memory and attention. This systematic review of randomized controlled trials (RCTs) aims to identify the effects of fruit and vegetable consumption on memory and attention. METHODS We conducted a systematic search in EBSCOhost, ProQuest, PubMed, Embase, and Web of Science from inception up to 06/09/2022. The inclusion criteria were peer-reviewed articles, fruit and vegetable intake measured using randomized controlled trials, and the outcome measures that showed the results of memory and attention scores. Two researchers independently extracted articles that met the selection criteria and evaluated the quality of each study. RESULTS There were 70 articles identified from the databases, of which 13 articles met the inclusion criteria and were included in this systematic review. There were 493 participants in total. The results show that consumption of fruit and vegetable intake improved memory and attention in longitudinal studies (10 to 12 weeks). Children showed improvement in immediate recall after supplementation with blueberries. Older adults required a higher dose of fruit and vegetable intake consumption to achieve significant improvement compared with children and younger adults. Furthermore, the effect of fruits and vegetables on memory showed better immediate memory recall than delayed recall. CONCLUSION This systematic review showed that there is an improvement in memory and attention with fruit and vegetable intake consumption. Hence, awareness of fruit and vegetable intake consumption is important to maintain cognitive health.
Collapse
Affiliation(s)
- Khai Ling Khor
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Selangor, Malaysia
- School of Psychology, DISTED College, Penang, Malaysia
| | - Vashnarekha Kumarasuriar
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Selangor, Malaysia
- School of Psychology and Clinical Language Sciences, University of Reading Malaysia, Iskandar Puteri, Malaysia
| | - Kok Wei Tan
- School of Psychology and Clinical Language Sciences, University of Reading Malaysia, Iskandar Puteri, Malaysia
| | - Pei Boon Ooi
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Selangor, Malaysia
| | - Yook-Chin Chia
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Selangor, Malaysia.
- Department of Primary Care Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
11
|
Nägele FL, Petersen M, Mayer C, Bönstrup M, Schulz R, Gerloff C, Thomalla G, Cheng B. Longitudinal microstructural alterations surrounding subcortical ischemic stroke lesions detected by free-water imaging. Hum Brain Mapp 2024; 45:e26722. [PMID: 38780442 PMCID: PMC11114091 DOI: 10.1002/hbm.26722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 03/20/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
In this study we explore the spatio-temporal trajectory and clinical relevance of microstructural white matter changes within and beyond subcortical stroke lesions detected by free-water imaging. Twenty-seven patients with subcortical infarct with mean age of 66.73 (SD 11.57) and median initial NIHSS score of 4 (IQR 3-7) received diffusion MRI 3-5 days, 1 month, 3 months, and 12 months after symptom-onset. Extracellular free-water and fractional anisotropy of the tissue (FAT) were averaged within stroke lesions and the surrounding tissue. Linear models showed increased free-water and decreased FAT in the white matter of patients with subcortical stroke (lesion [free-water/FAT, mean relative difference in %, ipsilesional vs. contralesional hemisphere at 3-5 days, 1 month, 3 months, and 12 months after symptom-onset]: +41/-34, +111/-37, +208/-26, +251/-18; perilesional tissue [range in %]: +[5-24]/-[0.2-7], +[2-20]/-[3-16], +[5-43]/-[2-16], +[10-110]/-[2-12]). Microstructural changes were most prominent within the lesion and gradually became less pronounced with increasing distance from the lesion. While free-water elevations continuously increased over time and peaked after 12 months, FAT decreases were most evident 1 month post-stroke, gradually returning to baseline values thereafter. Higher perilesional free-water and higher lesional FAT at baseline were correlated with greater reductions in lesion size (rho = -0.51, p = .03) in unadjusted analyses only, while there were no associations with clinical measures. In summary, we find a characteristic spatio-temporal pattern of extracellular and cellular alterations beyond subcortical stroke lesions, indicating a dynamic parenchymal response to ischemia characterized by vasogenic edema, cellular damage, and white matter atrophy.
Collapse
Affiliation(s)
- Felix L. Nägele
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Marvin Petersen
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Carola Mayer
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Marlene Bönstrup
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- Department of NeurologyUniversity of Leipzig Medical CenterLeipzigGermany
| | - Robert Schulz
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Christian Gerloff
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Götz Thomalla
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Bastian Cheng
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| |
Collapse
|
12
|
Li H, Cui F, Wang T, Wang W, Zhang D. The impact of sunlight exposure on brain structural markers in the UK Biobank. Sci Rep 2024; 14:10313. [PMID: 38705875 PMCID: PMC11070413 DOI: 10.1038/s41598-024-59633-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 04/12/2024] [Indexed: 05/07/2024] Open
Abstract
Sunlight is closely intertwined with daily life. It remains unclear whether there are associations between sunlight exposure and brain structural markers. General linear regression analysis was used to compare the differences in brain structural markers among different sunlight exposure time groups. Stratification analyses were performed based on sex, age, and diseases (hypertension, stroke, diabetes). Restricted cubic spline was performed to examine the dose-response relationship between natural sunlight exposure and brain structural markers, with further stratification by season. A negative association of sunlight exposure time with brain structural markers was found in the upper tertile compared to the lower tertile. Prolonged natural sunlight exposure was associated with the volumes of total brain (β: - 0.051, P < 0.001), white matter (β: - 0.031, P = 0.023), gray matter (β: - 0.067, P < 0.001), and white matter hyperintensities (β: 0.059, P < 0.001). These associations were more pronounced in males and individuals under the age of 60. The results of the restricted cubic spline analysis showed a nonlinear relationship between sunlight exposure and brain structural markers, with the direction changing around 2 h of sunlight exposure. This study demonstrates that prolonged exposure to natural sunlight is associated with brain structural markers change.
Collapse
Affiliation(s)
- Huihui Li
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 308 Ningxia Road, QingdaoShandong Province, 266071, China
| | - Fusheng Cui
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 308 Ningxia Road, QingdaoShandong Province, 266071, China
| | - Tong Wang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 308 Ningxia Road, QingdaoShandong Province, 266071, China
| | - Weijing Wang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 308 Ningxia Road, QingdaoShandong Province, 266071, China.
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, No. 308 Ningxia Road, QingdaoShandong Province, 266071, China.
| |
Collapse
|
13
|
Veeravalli KK. Implications of MMP-12 in the pathophysiology of ischaemic stroke. Stroke Vasc Neurol 2024; 9:97-107. [PMID: 37336584 PMCID: PMC11103161 DOI: 10.1136/svn-2023-002363] [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: 02/03/2023] [Accepted: 06/05/2023] [Indexed: 06/21/2023] Open
Abstract
This article focuses on the emerging role of matrix metalloproteinase-12 (MMP-12) in ischaemic stroke (IS). MMP-12 expression in the brain increases dramatically in animal models of IS, and its suppression reduces brain damage and promotes neurological, sensorimotor and cognitive functional outcomes. Thus, MMP-12 could represent a potential target for the management of IS. This article provides an overview of MMP-12 upregulation in the brain following IS, its deleterious role in the post-stroke pathogenesis (blood-brain barrier disruption, inflammation, apoptosis and demyelination), possible molecular interactions and mechanistic insights, its involvement in post-ischaemic functional deficits and recovery as well as the limitations, perspectives, challenges and future directions for further research. Prior to testing any MMP-12-targeted therapy in patients with acute IS, additional research is needed to establish the effectiveness of MMP-12 suppression against IS in older animals and in animals with comorbidities. This article also examines the clinical implications of suppressing MMP-12 alone or in combination with MMP-9 for extending the currently limited tissue plasminogen activator therapy time window. Targeting of MMP-12 is expected to have a profound influence on the therapeutic management of IS in the future.
Collapse
Affiliation(s)
- Krishna Kumar Veeravalli
- Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, USA
| |
Collapse
|
14
|
Parrilla GE, Gupta V, Wall RV, Salkar A, Basavarajappa D, Mirzaei M, Chitranshi N, Graham SL, You Y. The role of myelin in neurodegeneration: implications for drug targets and neuroprotection strategies. Rev Neurosci 2024; 35:271-292. [PMID: 37983528 DOI: 10.1515/revneuro-2023-0081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/27/2023] [Indexed: 11/22/2023]
Abstract
Myelination of axons in the central nervous system offers numerous advantages, including decreased energy expenditure for signal transmission and enhanced signal speed. The myelin sheaths surrounding an axon consist of a multi-layered membrane that is formed by oligodendrocytes, while specific glycoproteins and lipids play various roles in this formation process. As beneficial as myelin can be, its dysregulation and degeneration can prove detrimental. Inflammation, oxidative stress, and changes in cellular metabolism and the extracellular matrix can lead to demyelination of these axons. These factors are hallmark characteristics of certain demyelinating diseases including multiple sclerosis. The effects of demyelination are also implicated in primary degeneration in diseases such as glaucoma and Alzheimer's disease, as well as in processes of secondary degeneration. This reveals a relationship between myelin and secondary processes of neurodegeneration, including resultant degeneration following traumatic injury and transsynaptic degeneration. The role of myelin in primary and secondary degeneration is also of interest in the exploration of strategies and targets for remyelination, including the use of anti-inflammatory molecules or nanoparticles to deliver drugs. Although the use of these methods in animal models of diseases have shown to be effective in promoting remyelination, very few clinical trials in patients have met primary end points. This may be due to shortcomings or considerations that are not met while designing a clinical trial that targets remyelination. Potential solutions include diversifying disease targets and requiring concomitant interventions to promote rehabilitation.
Collapse
Affiliation(s)
- Gabriella E Parrilla
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Vivek Gupta
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Roshana Vander Wall
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Akanksha Salkar
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Devaraj Basavarajappa
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Mehdi Mirzaei
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Nitin Chitranshi
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Stuart L Graham
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
- Save Sight Institute, University of Sydney, 8 Macquarie St, Sydney, NSW 2000, Australia
| | - Yuyi You
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
- Save Sight Institute, University of Sydney, 8 Macquarie St, Sydney, NSW 2000, Australia
| |
Collapse
|
15
|
Xiao HH, Zhang FR, Li S, Guo FF, Hou JL, Wang SC, Yu J, Li XY, Yang HJ. Xinshubao tablet rescues cognitive dysfunction in a mouse model of vascular dementia: Involvement of neurogenesis and neuroinflammation. Biomed Pharmacother 2024; 172:116219. [PMID: 38310654 DOI: 10.1016/j.biopha.2024.116219] [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: 09/17/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/06/2024] Open
Abstract
Vascular dementia (VaD) represents a severe cognitive dysfunction syndrome closed linked to cardiovascular function. In the present study, we assessed the potential of Xinshubao tablet (XSB), a traditional Chinese prescription widely used for cardiovascular diseases, to mitigate neuropathological damage in a mouse model of VaD and elucidated the underlying mechanisms. Our findings revealed that oral administration of XSB rescued the cardiac dysfunction resulting from bilateral common carotid artery stenosis (BCAS), improved the cerebral blood flow (CBF) and cognitive function, reduced white matter injury, inhibited excessive microglial and astrocytic activation, stimulated hippocampal neurogenesis, and reduced neural apoptosis in the brains of BCAS mice. Mechanistically, RNA-seq analysis indicated that XSB treatment was significantly associated with neuroinflammation, vasculature development, and synaptic transmission, which were further confirmed by q-PCR assays. Western blot results revealed that XSB treatment hindered the nuclear translocation of nuclear factor-κB (NF-κB), thereby suppressing the NF-κB signaling pathway. These results collectively demonstrated that XSB could ameliorate cognitive dysfunction caused by BCAS through regulating CBF, reducing white matter lesions, suppressing glial activation, promoting neurogenesis, and mitigating neuroinflammation. Notably, the NF-κB signaling pathway emerged as a pivotal player in this mechanism.
Collapse
Affiliation(s)
- Hong-He Xiao
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Fujian Pien Tze Huang Enterprise Key Laboratory of Natural Medicine Research and Development, Zhangzhou Pien Tze Huang Pharmaceutical Co., Ltd., Zhangzhou, Fujian Province 363099, China
| | - Feng-Rong Zhang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Sen Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Fei-Fei Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jin-Li Hou
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Shi-Cong Wang
- Fujian Pien Tze Huang Enterprise Key Laboratory of Natural Medicine Research and Development, Zhangzhou Pien Tze Huang Pharmaceutical Co., Ltd., Zhangzhou, Fujian Province 363099, China
| | - Juan Yu
- Fujian Pien Tze Huang Enterprise Key Laboratory of Natural Medicine Research and Development, Zhangzhou Pien Tze Huang Pharmaceutical Co., Ltd., Zhangzhou, Fujian Province 363099, China.
| | - Xian-Yu Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Hong-Jun Yang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
16
|
Nunes GDF, Osso LA, Haynes JA, Morris A, Conant L, Stockton ME, Thornton MA, Vivian JA, Gandhi R, Denman DJ, Hughes EG. Incomplete remyelination via endogenous or therapeutically enhanced oligodendrogenesis is sufficient to recover visual cortical function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.21.581491. [PMID: 39411163 PMCID: PMC11475873 DOI: 10.1101/2024.02.21.581491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Myelin loss induces deficits in action potential propagation that result in neural dysfunction and contribute to the pathophysiology of neurodegenerative diseases, injury conditions, and aging. Because remyelination is often incomplete, better understanding endogenous remyelination and developing remyelination therapies that seek to restore neural function are clinical imperatives. Here, we used in vivo two-photon microscopy and electrophysiology to study the dynamics of endogenous and therapeutic-induced cortical remyelination and functional recovery after cuprizone-mediated demyelination in mice. We focused on the visual pathway, which is uniquely positioned to provide insights into structure-function relationships during de/remyelination. We show that endogenous remyelination is driven by recent oligodendrocyte loss and is highly efficacious following mild demyelination, but fails to restore the oligodendrocyte population when high rates of oligodendrocyte loss occur too quickly. Testing a novel thyromimetic compared to clemastine fumarate, we find it better enhances oligodendrocyte gain during remyelination and hastens recovery of neuronal function. Surprisingly, its therapeutic benefit was temporally restricted, and it acted exclusively following moderate to severe demyelination to eliminate endogenous remyelination deficits. However, complete remyelination is unnecessary as partial oligodendrocyte restoration was sufficient to recover visual neuronal function. These findings advance our understanding of remyelination and its impact on functional recovery to inform future therapeutic strategies.
Collapse
|
17
|
Deng X, Hu Z, Zhou S, Wu Y, Fu M, Zhou C, Sun J, Gao X, Huang Y. Perspective from single-cell sequencing: Is inflammation in acute ischemic stroke beneficial or detrimental? CNS Neurosci Ther 2024; 30:e14510. [PMID: 37905592 PMCID: PMC10805403 DOI: 10.1111/cns.14510] [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: 07/05/2023] [Revised: 09/24/2023] [Accepted: 10/08/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND Acute ischemic stroke (AIS) is a common cerebrovascular event associated with high incidence, disability, and poor prognosis. Studies have shown that various cell types, including microglia, astrocytes, oligodendrocytes, neurons, and neutrophils, play complex roles in the early stages of AIS and significantly affect its prognosis. Thus, a comprehensive understanding of the mechanisms of action of these cells will be beneficial for improving stroke prognosis. With the rapid development of single-cell sequencing technology, researchers have explored the pathophysiological mechanisms underlying AIS at the single-cell level. METHOD We systematically summarize the latest research on single-cell sequencing in AIS. RESULT In this review, we summarize the phenotypes and functions of microglia, astrocytes, oligodendrocytes, neurons, neutrophils, monocytes, and lymphocytes, as well as their respective subtypes, at different time points following AIS. In particular, we focused on the crosstalk between microglia and astrocytes, oligodendrocytes, and neurons. Our findings reveal diverse and sometimes opposing roles within the same cell type, with the possibility of interconversion between different subclusters. CONCLUSION This review offers a pioneering exploration of the functions of various glial cells and cell subclusters after AIS, shedding light on their regulatory mechanisms that facilitate the transformation of detrimental cell subclusters towards those that are beneficial for improving the prognosis of AIS. This approach has the potential to advance the discovery of new specific targets and the development of drugs, thus representing a significant breakthrough in addressing the challenges in AIS treatment.
Collapse
Affiliation(s)
- Xinpeng Deng
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Ziliang Hu
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Shengjun Zhou
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Yiwen Wu
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Menglin Fu
- School of Economics and ManagementChina University of GeosciencesWuhanChina
| | - Chenhui Zhou
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Jie Sun
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Xiang Gao
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Yi Huang
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| |
Collapse
|
18
|
Fan PL, Wang SS, Chu SF, Chen NH. Time-dependent dual effect of microglia in ischemic stroke. Neurochem Int 2023; 169:105584. [PMID: 37454817 DOI: 10.1016/j.neuint.2023.105584] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Stroke, the third leading cause of death and disability worldwide, is classified into ischemic or hemorrhagic, in which approximately 85% of strokes are ischemic. Ischemic stroke occurs as a result of arterial occlusion due to embolus or thrombus, with ischemia in the perfusion territory supplied by the occluded artery. The traditional concept that ischemic stroke is solely a vascular occlusion disorder has been expanded to include the dynamic interaction between microglia, astrocytes, neurons, vascular cells, and matrix components forming the "neurovascular unit." Acute ischemic stroke triggers a wide spectrum of neurovascular disturbances, glial activation, and secondary neuroinflammation that promotes further injury, ultimately resulting in neuronal death. Microglia, as the resident macrophages in the central nervous system, is one of the first responders to ischemic injury and plays a significant role in post-ischemic neuroinflammation. In this review, we reviewed the mechanisms of microglia in multiple stages of post-ischemic neuroinflammation development, including acute, sub-acute and chronic phases of stroke. A comprehensive understanding of the dynamic variation and the time-dependent role of microglia in post-stroke neuroinflammation could aid in the search for more effective therapeutics and diagnostic strategies for ischemic stroke.
Collapse
Affiliation(s)
- Ping-Long Fan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Sha-Sha Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Nai-Hong Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
19
|
Lu W, Wen J. H 2S-RhoA/ROCK Pathway and Glial Cells in Axonal Remyelination After Ischemic Stroke. Mol Neurobiol 2023; 60:5493-5504. [PMID: 37322287 DOI: 10.1007/s12035-023-03422-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 06/03/2023] [Indexed: 06/17/2023]
Abstract
Ischemic stroke is one of the main reasons of disability and death. Stroke-induced functional deficits are mainly due to the secondary degeneration of the white matter characterized by axonal demyelination and injury of axon-glial integrity. Enhancement of the axonal regeneration and remyelination could promote the neural functional recovery. However, cerebral ischemia-induced activation of RhoA/Rho kinase (ROCK) pathway plays a crucial and harmful role in the process of axonal recovery and regeneration. Inhibition of this pathway could promote the axonal regeneration and remyelination. In addition, hydrogen sulfide (H2S) has the significant neuroprotective role during the recovery of ischemic stroke via inhibiting the inflammatory response and oxidative stress, regulating astrocyte function, promoting the differentiation of endogenous oligodendrocyte precursor cells (OPCs) to mature oligodendrocyte. Among all of these effects, promoting the formation of mature oligodendrocyte is a crucial part of axonal regeneration and remyelination. Furthermore, numerous studies have uncovered the crosstalk between astrocytes and oligodendrocyte, microglial cells and oligodendrocyte in the axonal remyelination following ischemic stroke. The purpose of this review was to discuss the relationship among H2S, RhoA/ROCK pathway, astrocytes, and microglial cells in the axonal remyelination following ischemic stroke to reveal new strategies for preventing and treating this devastating disease.
Collapse
Affiliation(s)
- Weizhuo Lu
- Medical Branch, Hefei Technology College, Hefei, China
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
20
|
Jiang G, Ayaki T, Maki T, Yasuda K, Yoshii D, Kaji S, Takahashi R. Evaluation of BCAS1-positive immature oligodendrocytes after cerebral ischemic stroke and SVD. Neurosci Lett 2023; 812:137405. [PMID: 37479175 DOI: 10.1016/j.neulet.2023.137405] [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: 05/02/2023] [Revised: 07/02/2023] [Accepted: 07/18/2023] [Indexed: 07/23/2023]
Abstract
Ischemic cerebrovascular disease is an important cause of physical disability and dementia. Oligodendrocytes (OLGs), which differentiate from oligodendrocyte precursor cells (OPCs), are crucial for remyelination of the damaged brain and functional recovery. Breast carcinoma amplified sequence 1 (BCAS1) has recently been shown to be highly expressed in newly formed pre-myelinating oligodendrocytes (pre-mOLGs), while its expression level is reduced in mature OLGs. In this study, we analyzed BCAS1 expression by immunohistochemical analysis of human post-mortem brain tissue from six stroke patients (death within 2 months after stroke onset) and eight small vessel disease (SVD) patients. Control post-mortem brain tissue was from eight age-matched patients without any obvious central nervous system (CNS) pathology. The Olig2 expression in the area corresponding to the same section of the BCAS1-stained slice was analyzed to determine the total oligodendrocyte lineage. The percentage of differentiating OPCs in the oligodendrocyte lineage was calculated as the ratio of BCAS1+ to Olig2+ cells (BCAS1+/Olig2+). The stroke and SVD cases showed demyelination with decreased expression of myelin basic protein (MBP, a mature OLG marker). The stroke cases showed significantly increased numbers of early-stage BCAS1+ cells with an immature morphology and Olig2+ cells (pan-oligodendrocyte lineages) in the peri-infarct areas in both the cortex and white matter, but showed no increase in the number of late-stage BCAS1+ cells with a mature morphology. In contrast, the SVD cases showed no significant increase in Olig2+ and BCAS1+ cells. These results indicated that remyelination dysfunction could be attributed to insufficient maturation of OPCs in stroke and impaired recruitment of OPCs in SVD.
Collapse
Affiliation(s)
- Guanhua Jiang
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takashi Ayaki
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Takakuni Maki
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ken Yasuda
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Daisuke Yoshii
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Seiji Kaji
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
21
|
Martinez-Curiel R, Jansson L, Tsupykov O, Avaliani N, Aretio-Medina C, Hidalgo I, Monni E, Bengzon J, Skibo G, Lindvall O, Kokaia Z, Palma-Tortosa S. Oligodendrocytes in human induced pluripotent stem cell-derived cortical grafts remyelinate adult rat and human cortical neurons. Stem Cell Reports 2023; 18:1643-1656. [PMID: 37236198 PMCID: PMC10444570 DOI: 10.1016/j.stemcr.2023.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Neuronal loss and axonal demyelination underlie long-term functional impairments in patients affected by brain disorders such as ischemic stroke. Stem cell-based approaches reconstructing and remyelinating brain neural circuitry, leading to recovery, are highly warranted. Here, we demonstrate the in vitro and in vivo production of myelinating oligodendrocytes from a human induced pluripotent stem cell (iPSC)-derived long-term neuroepithelial stem (lt-NES) cell line, which also gives rise to neurons with the capacity to integrate into stroke-injured, adult rat cortical networks. Most importantly, the generated oligodendrocytes survive and form myelin-ensheathing human axons in the host tissue after grafting onto adult human cortical organotypic cultures. This lt-NES cell line is the first human stem cell source that, after intracerebral delivery, can repair both injured neural circuitries and demyelinated axons. Our findings provide supportive evidence for the potential future use of human iPSC-derived cell lines to promote effective clinical recovery following brain injuries.
Collapse
Affiliation(s)
- Raquel Martinez-Curiel
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Linda Jansson
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Oleg Tsupykov
- Department of Cytology, Bogomoletz Institute of Physiology; Institute of Genetic and Regenerative Medicine, Strazhesko National Scientific Center of Cardiology, Clinical and Regenerative Medicine, 01024 Kyiv, Ukraine
| | | | - Constanza Aretio-Medina
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Isabel Hidalgo
- Division of Molecular Hematology, Wallenberg Center for Molecular Medicine, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Emanuela Monni
- Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Johan Bengzon
- Division of Neurosurgery, Department of Clinical Sciences Lund, University Hospital, 22184 Lund, Sweden
| | - Galyna Skibo
- Department of Cytology, Bogomoletz Institute of Physiology; Institute of Genetic and Regenerative Medicine, Strazhesko National Scientific Center of Cardiology, Clinical and Regenerative Medicine, 01024 Kyiv, Ukraine
| | - Olle Lindvall
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Zaal Kokaia
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden.
| | - Sara Palma-Tortosa
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| |
Collapse
|
22
|
Huang S, Ren C, Luo Y, Ding Y, Ji X, Li S. New insights into the roles of oligodendrocytes regulation in ischemic stroke recovery. Neurobiol Dis 2023:106200. [PMID: 37321419 DOI: 10.1016/j.nbd.2023.106200] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/20/2023] [Accepted: 06/12/2023] [Indexed: 06/17/2023] Open
Abstract
Oligodendrocytes (OLs), the myelin-forming cells of the central nervous system, are integral to axonal integrity and function. Hypoxia-ischemia episodes can cause severe damage to these vulnerable cells through excitotoxicity, oxidative stress, inflammation, and mitochondrial dysfunction, leading to axonal dystrophy, neuronal dysfunction, and neurological impairments. OLs damage can result in demyelination and myelination disorders, severely impacting axonal function, structure, metabolism, and survival. Adult-onset stroke, periventricular leukomalacia, and post-stroke cognitive impairment primarily target OLs, making them a critical therapeutic target. Therapeutic strategies targeting OLs, myelin, and their receptors should be given more emphasis to attenuate ischemia injury and establish functional recovery after stroke. This review summarizes recent advances on the function of OLs in ischemic injury, as well as the present and emerging principles that serve as the foundation for protective strategies against OL deaths.
Collapse
Affiliation(s)
- Shuangfeng Huang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China; Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Changhong Ren
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China; Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yumin Luo
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China; Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University, Detroit, MI, USA
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China; Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Sijie Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China; Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
23
|
Gao B, Wang S, Li J, Han N, Ge H, Zhang G, Chang M. HMGB1, angel or devil, in ischemic stroke. Brain Behav 2023; 13:e2987. [PMID: 37062906 PMCID: PMC10176004 DOI: 10.1002/brb3.2987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/03/2023] [Accepted: 03/14/2023] [Indexed: 04/18/2023] Open
Abstract
INTRODUCTION High-mobility group box 1 protein (HMGB1) is extensively involved in causing ischemic stroke, pathological damage of ischemic brain injury, and neural tissue repair after ischemic brain injury. However, the precise role of HMGB1 in ischemic stroke remains to be elucidated. METHODS Comprehensive literature search and narrative review to summarize the current field of HMGB1 in cerebral ischemic based on the basic structure, structural modification, and functional roles of HMGB1 described in the literature. RESULTS Studies have exhibited the crucial roles of HMGB1 in cell death, immunity and inflammation, thrombosis, and remodeling and repair. HMGB1 released after cerebral infarction is extensively involved in the pathological injury process in the early stage of cerebral infarction, whereas it is involved in the promotion of brain tissue repair and remodeling in the late stage of cerebral infarction. HMGB1 plays a neurotrophic role in acute white matter stroke, whereas it causes sustained activation of inflammation and plays a damaging role in chronic white matter ischemia. CONCLUSIONS HMGB1 plays a complex role in cerebral infarction, which is related to not only the modification of HMGB1 and bound receptors but also different stages and subtypes of cerebral infarction. future studies on HMGB1 should investigate the spatial and temporal dynamics of HMGB1 after cerebral infarction. Moreover, future studies on HMGB1 should attempt to integrate different stages and infarct subtypes of cerebral infarction.
Collapse
Affiliation(s)
- Bin Gao
- Department of NeurologyXi'an No. 3 Hospitalthe Affiliated Hospital of Northest UniversityXi'anShaanxiP.R. China
| | - Shuwen Wang
- Department of NeurologyXi'an No. 3 Hospitalthe Affiliated Hospital of Northest UniversityXi'anShaanxiP.R. China
| | - Jiangfeng Li
- Department of Neurosurgerythe First Hospital of Yu'linYu'linShaanxiChina
| | - Nannan Han
- Department of NeurologyXi'an No. 3 Hospitalthe Affiliated Hospital of Northest UniversityXi'anShaanxiP.R. China
| | - Hanming Ge
- Department of NeurologyXi'an No. 3 Hospitalthe Affiliated Hospital of Northest UniversityXi'anShaanxiP.R. China
| | - Gejuan Zhang
- Department of NeurologyXi'an No. 3 Hospitalthe Affiliated Hospital of Northest UniversityXi'anShaanxiP.R. China
| | - Mingze Chang
- Department of NeurologyXi'an No. 3 Hospitalthe Affiliated Hospital of Northest UniversityXi'anShaanxiP.R. China
| |
Collapse
|
24
|
Xu T, Liu C, Deng S, Gan L, Zhang Z, Yang GY, Tian H, Tang Y. The roles of microglia and astrocytes in myelin phagocytosis in the central nervous system. J Cereb Blood Flow Metab 2023; 43:325-340. [PMID: 36324281 PMCID: PMC9941857 DOI: 10.1177/0271678x221137762] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
Myelination is an important process in the central nervous system (CNS). Oligodendrocytes (OLs) extend multiple layers to densely sheath on axons, composing the myelin to achieve efficient electrical signal conduction. The myelination during developmental stage maintains a balanced state. However, numerous CNS diseases including neurodegenerative and cerebrovascular diseases cause demyelination and disrupt the homeostasis, resulting in inflammation and white matter deficits. Effective clearance of myelin debris is needed in the region of demyelination, which is a key step for remyelination and tissue regeneration. Microglia and astrocytes are the major resident phagocytic cells in the brain, which may play different or collaborative roles in myelination. Microglia and astrocytes participate in developmental myelination through engulfing excessive unneeded myelin. They are also involved in the clearance of degenerated myelin debris for accelerating remyelination, or engulfing healthy myelin sheath for inhibiting remyelination. This review focuses on the roles of microglia and astrocytes in phagocytosing myelin in the developmental brain and diseased brain. In addition, the interaction between microglia and astrocytes to mediate myelin engulfment is also summarized.
Collapse
Affiliation(s)
- Tongtong Xu
- Shanghai Jiao Tong Affiliated Sixth People’s
Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University,
Shanghai, China
| | - Chang Liu
- Shanghai Jiao Tong Affiliated Sixth People’s
Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University,
Shanghai, China
| | - Shiyu Deng
- Shanghai Jiao Tong Affiliated Sixth People’s
Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University,
Shanghai, China
| | - Lin Gan
- Shanghai Jiao Tong Affiliated Sixth People’s
Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University,
Shanghai, China
| | - Zhijun Zhang
- Shanghai Jiao Tong Affiliated Sixth People’s
Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University,
Shanghai, China
| | - Guo-Yuan Yang
- Shanghai Jiao Tong Affiliated Sixth People’s
Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University,
Shanghai, China
| | - Hengli Tian
- Shanghai Jiao Tong Affiliated Sixth People’s
Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University,
Shanghai, China
| | - Yaohui Tang
- Shanghai Jiao Tong Affiliated Sixth People’s
Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University,
Shanghai, China
| |
Collapse
|
25
|
Wang H, Liu Y, Guo Z, Cui M, Pang P, Yang J, Wu C. Enhancement of oligodendrocyte autophagy alleviates white matter injury and cognitive impairment induced by chronic cerebral hypoperfusion in rats. Acta Pharm Sin B 2023; 13:2107-2123. [DOI: 10.1016/j.apsb.2023.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/23/2022] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
|
26
|
Yang Z, Gong M, Yang C, Chen C, Zhang K. Applications of Induced Pluripotent Stem Cell-Derived Glia in Brain Disease Research and Treatment. Handb Exp Pharmacol 2023; 281:103-140. [PMID: 37735301 DOI: 10.1007/164_2023_697] [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: 09/23/2023]
Abstract
Glia are integral components of neural networks and are crucial in both physiological functions and pathological processes of the brain. Many brain diseases involve glial abnormalities, including inflammatory changes, mitochondrial damage, calcium signaling disturbance, hemichannel opening, and loss of glutamate transporters. Induced pluripotent stem cell (iPSC)-derived glia provide opportunities to study the contributions of glia in human brain diseases. These cells have been used for human disease modeling as well as generating new therapies. This chapter introduces glial involvement in brain diseases, then summarizes different methods of generating iPSC-derived glia disease models of these cells. Finally, strategies for treating disease using iPSC-derived glia are discussed. The goal of this chapter is to provide an overview and shed light on the applications of iPSC-derived glia in brain disease research and treatment.
Collapse
Affiliation(s)
- Zhiqi Yang
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China
| | - Mingyue Gong
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China
| | - Chuanyan Yang
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China
| | - Chunhai Chen
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Kuan Zhang
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China.
| |
Collapse
|
27
|
Marzban M, Rustamzadeh A, Asghari A, Terme Y, Amichi AG, Ghanbarzehi V, Holaso AS, Hosseini F, Shahraki M, Sadafi P, Hashemzahi E, Honardar M, Iravankhah M, Baloochi M, Yarmohammadi A, Ebrahimi P. Stem cell therapy for cuprizone model of multiple sclerosis focusing on the effectiveness of different injection methods and cell labeling. Acta Histochem 2022; 124:151953. [PMID: 36116321 DOI: 10.1016/j.acthis.2022.151953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/06/2022] [Accepted: 09/12/2022] [Indexed: 11/01/2022]
Abstract
Multiple Sclerosis (MS) is a chronic and autoimmune disease of the central nervous system that causes inflammation in the brain and spinal cord, progressive degeneration of central nervous system tissue, damage to neuronal axons, and loss of function of central nervous system neurons. Experimental encephalomyelitis is an alternative animal model of MS that can simulate the symptoms of this disease. Cuprizone is one of the factors creating this model. Various researchers are testing the use of different cells to reduce the symptoms of cuprizone-demyelinated mice. The different injection methods explained in this article include intracerebral, intraperitoneal, intravenous, and intranasal. The intracerebral method, in contrast to the intranasal method, was widely employed by researchers. In each technique, the researchers try to inject a specific type of stem cell (SC) and monitor their efficiency. For monitoring SCs various labeling procedures are available, however, there is an upward trend in using magnetic resonance imaging (MRI). Two main barriers to using this method are high cost and complexity. In the current review, we try to make review cell therapy in the cuprizone model of MS.
Collapse
Affiliation(s)
- Mohsen Marzban
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran.
| | - Auob Rustamzadeh
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Aria Asghari
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Yousef Terme
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | | | - Vahid Ghanbarzehi
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | | | - Fateme Hosseini
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Mahya Shahraki
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Paniz Sadafi
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Erfan Hashemzahi
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Minoo Honardar
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Marziyeh Iravankhah
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Mehdi Baloochi
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Amin Yarmohammadi
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Pirooz Ebrahimi
- Department of Pharmacy, Health and Nutrition Sciences, University of Calabria, Italy
| |
Collapse
|
28
|
Higo N. Motor Cortex Plasticity During Functional Recovery Following Brain Damage. JOURNAL OF ROBOTICS AND MECHATRONICS 2022. [DOI: 10.20965/jrm.2022.p0700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Although brain damage causes functional impairment, it is often followed by partial or total recovery of function. Recovery is believed to occur primarily because of brain plasticity. Both human and animal studies have significantly contributed to uncovering the neuronal basis of plasticity. Recent advances in brain imaging technology have enabled the investigation of plastic changes in living human brains. In addition, animal experiments have revealed detailed changes at the neural and genetic levels. In this review, plasticity in motor-related areas of the cerebral cortex, which is one of the most well-studied areas of the neocortex in terms of plasticity, is reviewed. In addition, the potential of technological interventions to enhance plasticity and promote functional recovery following brain damage is discussed. Novel neurorehabilitation technologies are expected to be established based on the emerging research on plasticity from the last several decades.
Collapse
|
29
|
Chen D, Li J, Huang Y, Wei P, Miao W, Yang Y, Gao Y. Interleukin 13 promotes long-term recovery after ischemic stroke by inhibiting the activation of STAT3. J Neuroinflammation 2022; 19:112. [PMID: 35578342 PMCID: PMC9109418 DOI: 10.1186/s12974-022-02471-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/06/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Microglia/macrophages are activated after cerebral ischemic stroke and can contribute to either brain injury or recovery by polarizing microglia/macrophage into distinctive functional phenotypes with pro- or anti-inflammatory properties. Interleukin-13 (IL-13) is an anti-inflammatory cytokine that regulates microglia/macrophage polarization toward an anti-inflammatory phenotype. However, it is not clear whether IL-13 is beneficial after ischemic stroke long-term and the underlying molecular mechanism(s) remain unknown. Thus, we examined the effect of IL-13 on long-term recovery and microglia/macrophage polarization in mice with transient middle cerebral artery occlusion model (tMCAO). METHODS tMCAO was induced in adult male C57BL/6J mice. IL-13 (60 μg/kg) was administered intranasally starting 2 h after stroke and continued for seven consecutive days. Sensorimotor function, spatial learning and memory function, as well as brain infarct volume were assessed up to 35 days after stroke. White matter integrity was evaluated by electrophysiology, immunofluorescence staining, and transmission electron microscopy. Microglia/macrophage activation was assessed using immunofluorescence staining and quantitative real-time polymerase chain reaction. Changes in immune cells in the brain and the periphery, and expression of IL-13 receptors in different brain cells were detected by flow cytometry. Primary neuron/microglia co-cultures and a STAT3 inhibitor were used for mechanistic studies. RESULTS Post-treatment with IL-13 improved long-term neurofunctional recovery and decreased brain tissue atrophy after stroke. Intranasal delivery of IL-13 enhanced the structural and functional integrity of white matter after stroke. Furthermore, the neuroprotection afforded by IL-13 administration was not due to a direct effect on neurons, but by indirectly regulating the anti-inflammatory phenotype of microglia/macrophages. IL-13 treatment also had no effect on peripheral immune cells. Mechanistically, IL-13 improved the long-term outcome after ischemic stroke by promoting the polarization of microglia/macrophages toward the anti-inflammatory phenotype at least partially by inhibiting the phosphorylation of STAT3. CONCLUSIONS IL-13 promotes white matter repair and improves neurofunctional outcomes after ischemic stroke by modulating microglia/macrophages via inhibition of STAT3 phosphorylation.
Collapse
Affiliation(s)
- Di Chen
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, 200032, Shanghai, China
| | - Jiaying Li
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, 200032, Shanghai, China
| | - Yichen Huang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, 200032, Shanghai, China
| | - Pengju Wei
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, 200032, Shanghai, China
| | - Wanying Miao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, 200032, Shanghai, China
| | - Yaomei Yang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, 200032, Shanghai, China
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, 200032, Shanghai, China.
| |
Collapse
|
30
|
Wan T, Zhu W, Zhao Y, Zhang X, Ye R, Zuo M, Xu P, Huang Z, Zhang C, Xie Y, Liu X. Astrocytic phagocytosis contributes to demyelination after focal cortical ischemia in mice. Nat Commun 2022; 13:1134. [PMID: 35241660 PMCID: PMC8894352 DOI: 10.1038/s41467-022-28777-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 02/11/2022] [Indexed: 01/24/2023] Open
Abstract
Ischemic stroke can cause secondary myelin damage in the white matter distal to the primary injury site. The contribution of astrocytes during secondary demyelination and the underlying mechanisms are unclear. Here, using a mouse of distal middle cerebral artery occlusion, we show that lipocalin-2 (LCN2), enriched in reactive astrocytes, expression increases in nonischemic areas of the corpus callosum upon injury. LCN2-expressing astrocytes acquire a phagocytic phenotype and are able to uptake myelin. Myelin removal is impaired in Lcn2−/− astrocytes. Inducing re-expression of truncated LCN2(Δ2–20) in astrocytes restores phagocytosis and leads to progressive demyelination in Lcn2−/− mice. Co-immunoprecipitation experiments show that LCN2 binds to low-density lipoprotein receptor-related protein 1 (LRP1) in astrocytes. Knockdown of Lrp1 reduces LCN2-induced myelin engulfment by astrocytes and reduces demyelination. Altogether, our findings suggest that LCN2/LRP1 regulates astrocyte-mediated myelin phagocytosis in a mouse model of ischemic stroke. Ischemic stroke can cause secondary demyelination. Whether phagocytic astrocytes can contribute to such demyelination is unclear. Here, the authors show that lipocalin-2 (LCN-2) expression increased in astrocytes upon injury. LCN-2 expressing astrocytes acquire a phagocytic phenotype and contribute to secondary demyelination in a mouse model of ischemic stroke.
Collapse
Affiliation(s)
- Ting Wan
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China
| | - Wusheng Zhu
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China
| | - Ying Zhao
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China
| | - Xiaohao Zhang
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China
| | - Ruidong Ye
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China
| | - Meng Zuo
- Department of Neurology, Southwest Hospital and the First Affiliated Hospital, Army Medical University, Chongqing, 400000, China
| | - Pengfei Xu
- Stroke Center & Department of Neurology, The Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China
| | - Zhenqian Huang
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China
| | - Chunni Zhang
- Department of Clinical Laboratory, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China.
| | - Yi Xie
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China.
| | - Xinfeng Liu
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China. .,Stroke Center & Department of Neurology, The Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China.
| |
Collapse
|
31
|
Chourrout M, Rositi H, Ong E, Hubert V, Paccalet A, Foucault L, Autret A, Fayard B, Olivier C, Bolbos R, Peyrin F, Crola-da-Silva C, Meyronet D, Raineteau O, Elleaume H, Brun E, Chauveau F, Wiart M. Brain virtual histology with X-ray phase-contrast tomography Part I: whole-brain myelin mapping in white-matter injury models. BIOMEDICAL OPTICS EXPRESS 2022; 13:1620-1639. [PMID: 35415001 PMCID: PMC8973191 DOI: 10.1364/boe.438832] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/08/2021] [Accepted: 11/02/2021] [Indexed: 06/14/2023]
Abstract
White-matter injury leads to severe functional loss in many neurological diseases. Myelin staining on histological samples is the most common technique to investigate white-matter fibers. However, tissue processing and sectioning may affect the reliability of 3D volumetric assessments. The purpose of this study was to propose an approach that enables myelin fibers to be mapped in the whole rodent brain with microscopic resolution and without the need for strenuous staining. With this aim, we coupled in-line (propagation-based) X-ray phase-contrast tomography (XPCT) to ethanol-induced brain sample dehydration. We here provide the proof-of-concept that this approach enhances myelinated axons in rodent and human brain tissue. In addition, we demonstrated that white-matter injuries could be detected and quantified with this approach, using three animal models: ischemic stroke, premature birth and multiple sclerosis. Furthermore, in analogy to diffusion tensor imaging (DTI), we retrieved fiber directions and DTI-like diffusion metrics from our XPCT data to quantitatively characterize white-matter microstructure. Finally, we showed that this non-destructive approach was compatible with subsequent complementary brain sample analysis by conventional histology. In-line XPCT might thus become a novel gold-standard for investigating white-matter injury in the intact brain. This is Part I of a series of two articles reporting the value of in-line XPCT for virtual histology of the brain; Part II shows how in-line XPCT enables the whole-brain 3D morphometric analysis of amyloid- β (A β ) plaques.
Collapse
Affiliation(s)
- Matthieu Chourrout
- Univ-Lyon, Lyon Neuroscience
Research Center, CNRS UMR5292, Inserm U1028,
Université Claude Bernard Lyon 1, Lyon, France
- Co-first authors
| | - Hugo Rositi
- Univ-Clermont Auvergne; CNRS;
SIGMA Clermont; Institut Pascal,
Clermont-Ferrand, France
- Co-first authors
| | - Elodie Ong
- Univ-Lyon, CarMeN
laboratory, Inserm U1060, INRA U1397, Université
Claude Bernard Lyon 1, INSA Lyon, Charles Mérieux Medical
School, F-69600, Oullins, France
- Univ-Lyon, Hospices Civils de
Lyon, Lyon, France
| | - Violaine Hubert
- Univ-Lyon, CarMeN
laboratory, Inserm U1060, INRA U1397, Université
Claude Bernard Lyon 1, INSA Lyon, Charles Mérieux Medical
School, F-69600, Oullins, France
| | - Alexandre Paccalet
- Univ-Lyon, CarMeN
laboratory, Inserm U1060, INRA U1397, Université
Claude Bernard Lyon 1, INSA Lyon, Charles Mérieux Medical
School, F-69600, Oullins, France
| | - Louis Foucault
- Univ-Lyon, Université
Claude Bernard Lyon 1, Inserm, Stem Cell and Brain
Research Institute U1208, 69500 Bron, France
| | | | | | - Cécile Olivier
- Univ-Lyon, INSA-Lyon,
Université Claude Bernard Lyon 1,
CNRS, Inserm, CREATIS UMR5220, U1206, F-69621, France
| | | | - Françoise Peyrin
- Univ-Lyon, INSA-Lyon,
Université Claude Bernard Lyon 1,
CNRS, Inserm, CREATIS UMR5220, U1206, F-69621, France
| | - Claire Crola-da-Silva
- Univ-Lyon, CarMeN
laboratory, Inserm U1060, INRA U1397, Université
Claude Bernard Lyon 1, INSA Lyon, Charles Mérieux Medical
School, F-69600, Oullins, France
| | | | - Olivier Raineteau
- Univ-Lyon, Université
Claude Bernard Lyon 1, Inserm, Stem Cell and Brain
Research Institute U1208, 69500 Bron, France
| | - Héléne Elleaume
- Université Grenoble
Alpes, Inserm UA7 Strobe, Grenoble, France
| | - Emmanuel Brun
- Université Grenoble
Alpes, Inserm UA7 Strobe, Grenoble, France
| | - Fabien Chauveau
- Univ-Lyon, Lyon Neuroscience
Research Center, CNRS UMR5292, Inserm U1028,
Université Claude Bernard Lyon 1, Lyon, France
- CNRS, Lyon,
France
- Co-last authors
| | - Marlene Wiart
- Univ-Lyon, CarMeN
laboratory, Inserm U1060, INRA U1397, Université
Claude Bernard Lyon 1, INSA Lyon, Charles Mérieux Medical
School, F-69600, Oullins, France
- CNRS, Lyon,
France
- Co-last authors
| |
Collapse
|
32
|
Han B, Li X, Ai RS, Deng SY, Ye ZQ, Deng X, Ma W, Xiao S, Wang JZ, Wang LM, Xie C, Zhang Y, Xu Y, Zhang Y. Atmospheric particulate matter aggravates CNS demyelination through involvement of TLR-4/NF-kB signaling and microglial activation. eLife 2022; 11:72247. [PMID: 35199645 PMCID: PMC8893720 DOI: 10.7554/elife.72247] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 02/18/2022] [Indexed: 11/24/2022] Open
Abstract
Atmospheric Particulate Matter (PM) is one of the leading environmental risk factors for the global burden of disease. Increasing epidemiological studies demonstrated that PM plays a significant role in CNS demyelinating disorders; however, there is no direct testimony of this, and yet the molecular mechanism by which the occurrence remains unclear. Using multiple in vivo and in vitro strategies, in the present study we demonstrate that PM exposure aggravates neuroinflammation, myelin injury, and dysfunction of movement coordination ability via boosting microglial pro-inflammatory activities, in both the pathological demyelination and physiological myelinogenesis animal models. Indeed, pharmacological disturbance combined with RNA-seq and ChIP-seq suggests that TLR-4/NF-kB signaling mediated a core network of genes that control PM-triggered microglia pathogenicity. In summary, our study defines a novel atmospheric environmental mechanism that mediates PM-aggravated microglia pathogenic activities, and establishes a systematic approach for the investigation of the effects of environmental exposure in neurologic disorders.
Collapse
Affiliation(s)
- Bing Han
- Shaanxi Normal University, Xi'an, China
| | - Xing Li
- Shaanxi Normal University, Xi'an, China
| | | | | | | | - Xin Deng
- Shaanxi Normal University, Xi'an, China
| | - Wen Ma
- Shaanxi Normal University, Xi'an, China
| | - Shun Xiao
- Shaanxi Normal University, Xi'an, China
| | | | - Li-Mei Wang
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chong Xie
- Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yan Zhang
- Shaanxi Normal University, Xi'an, China
| | - Yan Xu
- Shaanxi Normal University, Xi'an, China
| | | |
Collapse
|
33
|
Huuskonen MT, Wang Y, Nikolakopoulou AM, Montagne A, Dai Z, Lazic D, Sagare AP, Zhao Z, Fernandez JA, Griffin JH, Zlokovic BV. Protection of ischemic white matter and oligodendrocytes in mice by 3K3A-activated protein C. J Exp Med 2022; 219:e20211372. [PMID: 34846535 PMCID: PMC8635278 DOI: 10.1084/jem.20211372] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/19/2021] [Accepted: 10/29/2021] [Indexed: 12/13/2022] Open
Abstract
Subcortical white matter (WM) stroke accounts for 25% of all strokes and is the second leading cause of dementia. Despite such clinical importance, we still do not have an effective treatment for ischemic WM stroke, and the mechanisms of WM postischemic neuroprotection remain elusive. 3K3A-activated protein C (APC) is a signaling-selective analogue of endogenous blood protease APC that is currently in development as a neuroprotectant for ischemic stroke patients. Here, we show that 3K3A-APC protects WM tracts and oligodendrocytes from ischemic injury in the corpus callosum in middle-aged mice by activating protease-activated receptor 1 (PAR1) and PAR3. We show that PAR1 and PAR3 were also required for 3K3A-APC's suppression of post-WM stroke microglia and astrocyte responses and overall improvement in neuropathologic and functional outcomes. Our data provide new insights into the neuroprotective APC pathway in the WM and illustrate 3K3A-APC's potential for treating WM stroke in humans, possibly including multiple WM strokes that result in vascular dementia.
Collapse
Affiliation(s)
- Mikko T. Huuskonen
- Department of Physiology and Neuroscience, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Yaoming Wang
- Department of Physiology and Neuroscience, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Angeliki Maria Nikolakopoulou
- Department of Physiology and Neuroscience, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Axel Montagne
- Department of Physiology and Neuroscience, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Zhonghua Dai
- Department of Physiology and Neuroscience, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Divna Lazic
- Department of Physiology and Neuroscience, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Abhay P. Sagare
- Department of Physiology and Neuroscience, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Zhen Zhao
- Department of Physiology and Neuroscience, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Jose A. Fernandez
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - John H. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
- Division of Hematology/Oncology, Department of Medicine, University of California, San Diego, San Diego, CA
| | - Berislav V. Zlokovic
- The Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| |
Collapse
|
34
|
Higo N. Non-human Primate Models to Explore the Adaptive Mechanisms After Stroke. Front Syst Neurosci 2021; 15:760311. [PMID: 34819842 PMCID: PMC8606408 DOI: 10.3389/fnsys.2021.760311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/20/2021] [Indexed: 01/15/2023] Open
Abstract
The brain has the ability to reconstruct neural structures and functions to compensate for the brain lesions caused by stroke, although it is highly limited in primates including humans. Animal studies in which experimental lesions were induced in the brain have contributed to the current understanding of the neural mechanisms underlying functional recovery. Here, I have highlighted recent advances in non-human primate models using primate species such as macaques and marmosets, most of which have been developed to study the mechanisms underlying the recovery of motor functions after stroke. Cortical lesion models have been used to investigate motor recovery after lesions to the cortical areas involved in movements of specific body parts. Models of a focal stroke at the posterior internal capsule have also been developed to bridge the gap between the knowledge obtained by cortical lesion models and the development of intervention strategies because the severity and outcome of motor deficits depend on the degree of lesions to the region. This review will also introduce other stroke models designed to study the plastic changes associated with development and recovery from cognitive and sensory impairments. Although further validation and careful interpretation are required, considering the differences between non-human primate brains and human brains, studies using brain-lesioned non-human primates offer promise for improving translational outcomes.
Collapse
Affiliation(s)
- Noriyuki Higo
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| |
Collapse
|
35
|
Khodanovich MY, Gubskiy IL, Kudabaeva MS, Namestnikova DD, Kisel AA, Anan’ina TV, Tumentceva YA, Mustafina LR, Yarnykh VL. Long-term monitoring of chronic demyelination and remyelination in a rat ischemic stroke model using macromolecular proton fraction mapping. J Cereb Blood Flow Metab 2021; 41:2856-2869. [PMID: 34107787 PMCID: PMC8756474 DOI: 10.1177/0271678x211020860] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 12/23/2022]
Abstract
Remyelination is a key process enabling post-stroke brain tissue recovery and plasticity. This study aimed to explore the feasibility of demyelination and remyelination monitoring in experimental stroke from the acute to chronic stage using an emerging myelin imaging biomarker, macromolecular proton fraction (MPF). After stroke induction by transient middle cerebral artery occlusion, rats underwent repeated MRI examinations during 85 days after surgery with histological endpoints for the animal subgroups on the 7th, 21st, 56th, and 85th days. MPF maps revealed two sub-regions within the infarct characterized by distinct temporal profiles exhibiting either a persistent decrease by 30%-40% or a transient decrease followed by return to nearly normal values after one month of observation. Myelin histology confirmed that these sub-regions had nearly similar extent of demyelination in the sub-acute phase and then demonstrated either chronic demyelination or remyelination. The remyelination zones also exhibited active axonal regrowth, reconstitution of compact fiber bundles, and proliferation of neuronal and oligodendroglial precursors. The demyelination zones showed more extensive astrogliosis from the 21st day endpoint. Both sub-regions had substantially depleted neuronal population over all endpoints. These results histologically validate MPF mapping as a novel approach for quantitative assessment of myelin damage and repair in ischemic stroke.
Collapse
Affiliation(s)
| | - Ilya L Gubskiy
- Research Institute of Cerebrovascular Pathology and Stroke, Pirogov Russian Medical University, Moscow, Russian Federation
| | - Marina S Kudabaeva
- Laboratory of Neurobiology, Tomsk State University, Tomsk, Russian Federation
| | - Darya D Namestnikova
- Research Institute of Cerebrovascular Pathology and Stroke, Pirogov Russian Medical University, Moscow, Russian Federation
| | - Alena A Kisel
- Laboratory of Neurobiology, Tomsk State University, Tomsk, Russian Federation
- Department of Radiology, University of Washington, Seattle, USA
| | - Tatyana V Anan’ina
- Laboratory of Neurobiology, Tomsk State University, Tomsk, Russian Federation
| | - Yana A Tumentceva
- Laboratory of Neurobiology, Tomsk State University, Tomsk, Russian Federation
| | - Lilia R Mustafina
- Department of histology, embriology, and cytology, Siberian State Medical University, Tomsk, Russian Federation
| | - Vasily L Yarnykh
- Laboratory of Neurobiology, Tomsk State University, Tomsk, Russian Federation
- Department of Radiology, University of Washington, Seattle, USA
| |
Collapse
|
36
|
Inhibition of the Immunoproteasome Subunit LMP7 Ameliorates Cerebral White Matter Demyelination Possibly via TGF β/Smad Signaling. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6426225. [PMID: 34675988 PMCID: PMC8526201 DOI: 10.1155/2021/6426225] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 08/12/2021] [Accepted: 09/13/2021] [Indexed: 12/03/2022]
Abstract
Objectives Chronic cerebral hypoperfusion induces white matter ischemic injury and cognitive impairment, whereas the mechanism remains unclear. Immunoproteasomes have been implicated in the pathogenesis of acute ischemia stroke and multiple sclerosis. However, the expression and role of immunoproteasomes in the brain of chronic cerebral hypoperfusion remain to be clarified. Methods Chronic white matter ischemic injury mice models were induced by bilateral carotid artery stenosis (BCAS). A selective immunoproteasome subunit low-molecular-mass peptide-7 (LMP7) inhibitor PR957 was administered to mice. Cognitive function, white matter integrity, and potential pathways were assessed after BCAS. Results The present study found that chronic cerebral hypoperfusion following BCAS induced cerebral white matter demyelination and cognitive impairment, accompanied with elevated expression of the immunoproteasomes LMP2 and LMP7, activation of astrocytes and microglia, and increased production of inflammatory cytokines (e.g., interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), IL-10, transforming growth factor-β1 (TGFβ1), and insulin-like growth factor-1 (IGF-1)). However, inhibition of LMP7 with the specific proteasome inhibitor PR957 significantly mitigated the histological damage of the white matter, suppressed inflammatory response, and paralleled by an improvement of cognitive function. Furthermore, treatment of PR957 significantly upregulated the level of TGFβ1, the total expression level, and the phosphorylation level of Smad2/3 and promoted brain remyelination. Surprisingly, PR957 alone had no effects on the neuroinflammation response and the activation of TGFβ/Smad signaling in the sham-operated (BCAS-nonoperated) mice. Conclusions The possible mechanism underlying this was attributed to that the immunoproteasome regulates TGFβ/Smad signaling-mediated neuroinflammation and oligodendrocyte remyelination.
Collapse
|
37
|
Curcumin Ameliorates White Matter Injury after Ischemic Stroke by Inhibiting Microglia/Macrophage Pyroptosis through NF- κB Suppression and NLRP3 Inflammasome Inhibition. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1552127. [PMID: 34630845 PMCID: PMC8497115 DOI: 10.1155/2021/1552127] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/25/2021] [Accepted: 09/01/2021] [Indexed: 12/19/2022]
Abstract
NLRP3 inflammasome-mediated pyroptosis is a proinflammatory programmed cell death pathway, which plays a vital role in functional outcomes after stroke. We previously described the beneficial effects of curcumin against stroke-induced neuronal damage through modulating microglial polarization. However, the impact of curcumin on microglial pyroptosis remains unknown. Here, stroke was modeled in mice by middle cerebral artery occlusion (MCAO) for 60 minutes and treated with curcumin (150 mg/kg) intraperitoneally immediately after reperfusion, followed by daily administrations for 7 days. Curcumin ameliorated white matter (WM) lesions and brain tissue loss 21 days poststroke and improved sensorimotor function 3, 10, and 21 days after stroke. Furthermore, curcumin significantly reduced the number of gasdermin D+ (GSDMD+) Iba1+ and caspase-1+Iba1+ microglia/macrophage 21 days after stroke. In vitro, lipopolysaccharide (LPS) with ATP treatment was used to induce pyroptosis in primary microglia. Western blot revealed a decrease in pyroptosis-related proteins, e.g., GSDMD-N, cleaved caspase-1, NLRP3, IL-1β, and IL-18, following in vitro or in vivo curcumin treatment. Mechanistically, both in vivo and in vitro studies confirmed that curcumin inhibited the activation of the NF-κB pathway. NLRP3 knocked down by siRNA transfection markedly increased the inhibitory effects of curcumin on microglial pyroptosis and proinflammatory responses, both in vitro and in vivo. Furthermore, stereotaxic microinjection of AAV-based NLRP3 shRNA significantly improved sensorimotor function and reduced WM lesion following curcumin treatment in MCAO mice. Our study suggested that curcumin reduced stroke-induced WM damage, improved functional outcomes, and attenuated microglial pyroptosis, at least partially, through suppression of the NF-κB/NLRP3 signaling pathway, further supporting curcumin as a potential therapeutic drug for stroke.
Collapse
|
38
|
Zhang J, Zheng W, Shang D, Chen Y, Zhong S, Ye J, Li L, Yu Y, Zhang L, Cheng R, He F, Wu D, Ye X, Luo B. Fixel-based evidence of microstructural damage in crossing pathways improves language mapping in Post-stroke aphasia. NEUROIMAGE-CLINICAL 2021; 31:102774. [PMID: 34371239 PMCID: PMC8358698 DOI: 10.1016/j.nicl.2021.102774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 07/16/2021] [Accepted: 07/22/2021] [Indexed: 12/28/2022]
Abstract
FBA shows greater specificity in mapping injured fibers in post-stroke aphasia. Intra-axonal volume of axons in dual streams is reduced in post-stroke aphasia. FBA could locate more precise segments associated with language defects. FBA could attribute language subdomain scores to fiber density of distinct tracts.
Background The complex crossing-fiber characteristics in the dual-stream system have been ignored by traditional diffusion tensor models regarding disconnections in post-stroke aphasia. It is valuable to identify microstructural damage of crossing-fiber pathways and reveal accurate fiber-specific language mapping in patients with aphasia. Methods This cross-sectional study collected magnetic resonance imaging data from 29 participants with post-stroke aphasia in the subacute stage and from 33 age- and sex-matched healthy controls. Fixel-based analysis was performed to examine microstructural fiber density (FD) and bundle cross-section alterations of specific fiber populations in crossing-fiber regions. Group comparisons were performed, and relationships with language scores were assessed. Results The aphasic group exhibited significant fixel-wise FD reductions in the dual-stream tracts, including the left inferior fronto-occipital fasciculus (IFOF), arcuate fasciculus, and superior longitudinal fasciculus (SLF) III (family-wise-error-corrected p < 0.05). Voxel- and fixel-wise comparisons revealed mismatched distributions in regions with crossing-fiber nexuses. Fixel-wise correlation analyses revealed significant associations between comprehension impairment and reduced FD in the temporal and frontal segments of the left IFOF, and also mapped naming ability to the IFOF. Average features along the whole course of dominant tracts assessed with tract-wise analyses attributed word-level comprehension to the IFOF (r = 0.723, p < 0.001) and revealed a trend-level correlation between sentence-level comprehension and FD of the SLF III (r = 0.451, p = 0.021). The mean FD of the uncinate fasciculus (UF) and IFOF correlated with total and picture naming scores, and the IFOF also correlated with responsive naming subdomains (Bonferroni corrected p < 0.05). Conclusions FD reductions of dual streams suggest that intra-axonal volume reduction constitutes the microstructural damage of white matter integrity in post-stroke aphasia. Fixel-based analysis provides a complementary method of language mapping that identifies fiber-specific tracts in the left hemisphere language network with greater specificity than voxel-based analysis. It precisely locates the precise segments of the IFOF for comprehension, yields fiber-specific evidence for the debated UF-naming association, and reveals dissociative subdomain associations with distinct tracts.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Neurology & Brain Medical Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Rehabilitation Medicine Center & Rehabilitation Research Institute of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Weihao Zheng
- School of Information Science and Engineering, Lanzhou University, Lanzhou, China
| | - Desheng Shang
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yating Chen
- Rehabilitation Medicine Center & Rehabilitation Research Institute of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Shuchang Zhong
- Rehabilitation Medicine Center & Rehabilitation Research Institute of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Jing Ye
- Rehabilitation Medicine Center & Rehabilitation Research Institute of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Lingling Li
- Department of Neurology & Brain Medical Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yamei Yu
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Li Zhang
- Rehabilitation Medicine Center & Rehabilitation Research Institute of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Ruidong Cheng
- Rehabilitation Medicine Center & Rehabilitation Research Institute of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Fangping He
- Department of Neurology & Brain Medical Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Wu
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Xiangming Ye
- Rehabilitation Medicine Center & Rehabilitation Research Institute of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Benyan Luo
- Department of Neurology & Brain Medical Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, China.
| |
Collapse
|
39
|
Mike JK, Ferriero DM. Efferocytosis Mediated Modulation of Injury after Neonatal Brain Hypoxia-Ischemia. Cells 2021; 10:1025. [PMID: 33925299 PMCID: PMC8146813 DOI: 10.3390/cells10051025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023] Open
Abstract
Neonatal brain hypoxia-ischemia (HI) is a leading cause of morbidity and long-term disabilities in children. While we have made significant progress in describing HI mechanisms, the limited therapies currently offered for HI treatment in the clinical setting stress the importance of discovering new targetable pathways. Efferocytosis is an immunoregulatory and homeostatic process of clearance of apoptotic cells (AC) and cellular debris, best described in the brain during neurodevelopment. The therapeutic potential of stimulating defective efferocytosis has been recognized in neurodegenerative diseases. In this review, we will explore the involvement of efferocytosis after a stroke and HI as a promising target for new HI therapies.
Collapse
Affiliation(s)
- Jana Krystofova Mike
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA;
| | - Donna Marie Ferriero
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA;
- Department of Neurology Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
40
|
Allan KC, Hu LR, Scavuzzo MA, Morton AR, Gevorgyan AS, Cohn EF, Clayton BL, Bederman IR, Hung S, Bartels CF, Madhavan M, Tesar PJ. Non-canonical Targets of HIF1a Impair Oligodendrocyte Progenitor Cell Function. Cell Stem Cell 2021; 28:257-272.e11. [PMID: 33091368 PMCID: PMC7867598 DOI: 10.1016/j.stem.2020.09.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/19/2020] [Accepted: 09/29/2020] [Indexed: 12/25/2022]
Abstract
Mammalian cells respond to insufficient oxygen through transcriptional regulators called hypoxia-inducible factors (HIFs). Although transiently protective, prolonged HIF activity drives distinct pathological responses in different tissues. Using a model of chronic HIF1a accumulation in pluripotent-stem-cell-derived oligodendrocyte progenitors (OPCs), we demonstrate that HIF1a activates non-canonical targets to impair generation of oligodendrocytes from OPCs. HIF1a activated a unique set of genes in OPCs through interaction with the OPC-specific transcription factor OLIG2. Non-canonical targets, including Ascl2 and Dlx3, were sufficient to block differentiation through suppression of the oligodendrocyte regulator Sox10. Chemical screening revealed that inhibition of MEK/ERK signaling overcame the HIF1a-mediated block in oligodendrocyte generation by restoring Sox10 expression without affecting canonical HIF1a activity. MEK/ERK inhibition also drove oligodendrocyte formation in hypoxic regions of human oligocortical spheroids. This work defines mechanisms by which HIF1a impairs oligodendrocyte formation and establishes that cell-type-specific HIF1a targets perturb cell function in response to low oxygen.
Collapse
Affiliation(s)
- Kevin C. Allan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Lucille R. Hu
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Marissa A. Scavuzzo
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Andrew R. Morton
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Artur S. Gevorgyan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Erin F. Cohn
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Benjamin L.L. Clayton
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Ilya R. Bederman
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Stevephen Hung
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Cynthia F. Bartels
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Mayur Madhavan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Paul J. Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA.,Lead Contact,Correspondence:
| |
Collapse
|
41
|
Cellular and Molecular Mechanisms of R/S-Roscovitine and CDKs Related Inhibition under Both Focal and Global Cerebral Ischemia: A Focus on Neurovascular Unit and Immune Cells. Cells 2021; 10:cells10010104. [PMID: 33429982 PMCID: PMC7827530 DOI: 10.3390/cells10010104] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/29/2020] [Accepted: 01/05/2021] [Indexed: 12/29/2022] Open
Abstract
Ischemic stroke is the second leading cause of death worldwide. Following ischemic stroke, Neurovascular Unit (NVU) inflammation and peripheral leucocytes infiltration are major contributors to the extension of brain lesions. For a long time restricted to neurons, the 10 past years have shown the emergence of an increasing number of studies focusing on the role of Cyclin-Dependent Kinases (CDKs) on the other cells of NVU, as well as on the leucocytes. The most widely used CDKs inhibitor, (R)-roscovitine, and its (S) isomer both decreased brain lesions in models of global and focal cerebral ischemia. We previously showed that (S)-roscovitine acted, at least, by modulating NVU response to ischemia. Interestingly, roscovitine was shown to decrease leucocytes-mediated inflammation in several inflammatory models. Specific inhibition of roscovitine majors target CDK 1, 2, 5, 7, and 9 showed that these CDKs played key roles in inflammatory processes of NVU cells and leucocytes after brain lesions, including ischemic stroke. The data summarized here support the investigation of roscovitine as a potential therapeutic agent for the treatment of ischemic stroke, and provide an overview of CDK 1, 2, 5, 7, and 9 functions in brain cells and leucocytes during cerebral ischemia.
Collapse
|
42
|
Age-related injury responses of human oligodendrocytes to metabolic insults: link to BCL-2 and autophagy pathways. Commun Biol 2021; 4:20. [PMID: 33398046 PMCID: PMC7782481 DOI: 10.1038/s42003-020-01557-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 12/01/2020] [Indexed: 01/29/2023] Open
Abstract
Myelin destruction and oligodendrocyte (OL) death consequent to metabolic stress is a feature of CNS disorders across the age spectrum. Using cells derived from surgically resected tissue, we demonstrate that young (<age 5) pediatric-aged sample OLs are more resistant to in-vitro metabolic injury than fetal O4+ progenitor cells, but more susceptible to cell death and apoptosis than adult-derived OLs. Pediatric but not adult OLs show measurable levels of TUNEL+ cells, a feature of the fetal cell response. The ratio of anti- vs pro-apoptotic BCL-2 family genes are increased in adult vs pediatric (<age 5) mature OLs and in more mature OL lineage cells. Lysosomal gene expression was increased in adult and pediatric compared to fetal OL lineage cells. Cell death of OLs was increased by inhibiting pro-apoptotic BCL-2 gene and autophagy activity. These distinct age-related injury responses should be considered in designing therapies aimed at reducing myelin injury.
Collapse
|
43
|
Nowack L, Teschers CS, Albrecht S, Gilmour R. Oligodendroglial glycolipids in (Re)myelination: implications for multiple sclerosis research. Nat Prod Rep 2021; 38:890-904. [PMID: 33575689 DOI: 10.1039/d0np00093k] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Covering: up to 2020 This short review surveys aspects of glycolipid-based natural products and their biological relevance in multiple sclerosis (MS). The role of isolated gangliosides in disease models is discussed together with an overview of ganglioside-inspired small molecule drugs and imaging probes. The discussion is extended to neurodegeneration in a more general context and addresses the need for more efficient synthetic methods to generate (glyco)structures that are of therapeutic relevance.
Collapse
Affiliation(s)
- Luise Nowack
- Institute for Organic Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstraße 36, 48149 Münster, Germany. and Institute of Neuropathology, University Hospital Münster, Pottkamp 2, 48149 Münster, Germany.
| | - Charlotte S Teschers
- Institute for Organic Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstraße 36, 48149 Münster, Germany.
| | - Stefanie Albrecht
- Institute of Neuropathology, University Hospital Münster, Pottkamp 2, 48149 Münster, Germany.
| | - Ryan Gilmour
- Institute for Organic Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstraße 36, 48149 Münster, Germany.
| |
Collapse
|
44
|
Chen D, Huang Y, Shi Z, Li J, Zhang Y, Wang K, Smith AD, Gong Y, Gao Y. Demyelinating processes in aging and stroke in the central nervous system and the prospect of treatment strategy. CNS Neurosci Ther 2020; 26:1219-1229. [PMID: 33210839 PMCID: PMC7702227 DOI: 10.1111/cns.13497] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/26/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
Demyelination occurs in response to brain injury and is observed in many neurodegenerative diseases. Myelin is synthesized from oligodendrocytes in the central nervous system, and oligodendrocyte death‐induced demyelination is one of the mechanisms involved in white matter damage after stroke and neurodegeneration. Oligodendrocyte precursor cells (OPCs) exist in the brain of normal adults, and their differentiation into mature oligodendrocytes play a central role in remyelination. Although the differentiation and maturity of OPCs drive endogenous efforts for remyelination, the failure of axons to remyelinate is still the biggest obstacle to brain repair after injury or diseases. In recent years, studies have made attempts to promote remyelination after brain injury and disease, but its cellular or molecular mechanism is not yet fully understood. In this review, we discuss recent studies examining the demyelination process and potential therapeutic strategies for remyelination in aging and stroke. Based on our current understanding of the cellular and molecular mechanisms underlying remyelination, we hypothesize that myelin and oligodendrocytes are viable therapeutic targets to mitigate brain injury and to treat demyelinating‐related neurodegeneration diseases.
Collapse
Affiliation(s)
- Di Chen
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yichen Huang
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ziyu Shi
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jiaying Li
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yue Zhang
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ke Wang
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Amanda D Smith
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Ye Gong
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yanqin Gao
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
45
|
Low-Field Magnetic Stimulation Accelerates the Differentiation of Oligodendrocyte Precursor Cells via Non-canonical TGF-β Signaling Pathways. Mol Neurobiol 2020; 58:855-866. [PMID: 33037982 DOI: 10.1007/s12035-020-02157-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/30/2020] [Indexed: 01/17/2023]
Abstract
Demyelination and oligodendrocyte loss are characteristic changes in demyelinating disorders. Low-field magnetic stimulation (LFMS) is a novel transcranial neuromodulation technology that has shown promising therapeutic potential for a variety of neuropsychiatric conditions. The cellular and molecular mechanisms of magnetic stimulation remain unclear. Previous studies mainly focused on the effects of magnetic stimulation on neuronal cells. Here we aimed to examine the effects of a gamma frequency LFMS on the glial progenitor cells. We used rat central glia-4 (CG4) cell line as an in vitro model. CG4 is a bipotential glial progenitor cell line that can differentiate into either oligodendrocyte or type 2-astrocyte. The cells cultured in a defined differentiation media were exposed to a 40-Hz LFMS 20 min daily for five consecutive days. We found that LFMS transiently elevated the level of TGF-β1 in the culture media in the first 24 h after the treatment. In correlation with the TGF-β1 levels, the percentage of cells possessing complex branches and expressing the late oligodendrocyte progenitor marker O4 was increased, indicating the accelerated differentiation of CG4 cells towards oligodendrocyte in LFMS-treated cultures. LFMS increased phosphorylation of Akt and Erk1/2 proteins, but not SMAD2/3. TGF-β1 receptor I specific inhibitor LY 364947 partially suppressed the effects of LFMS on differentiation and on levels of pAkt and pErk1/2, indicating that LFMS enhances the differentiation of oligodendrocyte progenitor cells via activation of non-canonical TGF-β-Akt and TGF-β-Erk1/2 pathways but not the canonical SMAD pathway. The data from this study reveal a novel mechanism of magnetic stimulation as a potential therapy for demyelination disorders.
Collapse
|
46
|
Melià-Sorolla M, Castaño C, DeGregorio-Rocasolano N, Rodríguez-Esparragoza L, Dávalos A, Martí-Sistac O, Gasull T. Relevance of Porcine Stroke Models to Bridge the Gap from Pre-Clinical Findings to Clinical Implementation. Int J Mol Sci 2020; 21:ijms21186568. [PMID: 32911769 PMCID: PMC7555414 DOI: 10.3390/ijms21186568] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
In the search of animal stroke models providing translational advantages for biomedical research, pigs are large mammals with interesting brain characteristics and wide social acceptance. Compared to rodents, pigs have human-like highly gyrencephalic brains. In addition, increasingly through phylogeny, animals have more sophisticated white matter connectivity; thus, ratios of white-to-gray matter in humans and pigs are higher than in rodents. Swine models provide the opportunity to study the effect of stroke with emphasis on white matter damage and neuroanatomical changes in connectivity, and their pathophysiological correlate. In addition, the subarachnoid space surrounding the swine brain resembles that of humans. This allows the accumulation of blood and clots in subarachnoid hemorrhage models mimicking the clinical condition. The clot accumulation has been reported to mediate pathological mechanisms known to contribute to infarct progression and final damage in stroke patients. Importantly, swine allows trustworthy tracking of brain damage evolution using the same non-invasive multimodal imaging sequences used in the clinical practice. Moreover, several models of comorbidities and pathologies usually found in stroke patients have recently been established in swine. We review here ischemic and hemorrhagic stroke models reported so far in pigs. The advantages and limitations of each model are also discussed.
Collapse
Affiliation(s)
- Marc Melià-Sorolla
- Cellular and Molecular Neurobiology Research Group, Department of Neurosciences, Germans Trias i Pujol Research Institute, 08916 Badalona, Catalonia, Spain; (M.M.-S.); (N.D.-R.)
| | - Carlos Castaño
- Neurointerventional Radiology Unit, Department of Neurosciences, Hospital Germans Trias i Pujol, 08916 Badalona, Catalonia, Spain;
| | - Núria DeGregorio-Rocasolano
- Cellular and Molecular Neurobiology Research Group, Department of Neurosciences, Germans Trias i Pujol Research Institute, 08916 Badalona, Catalonia, Spain; (M.M.-S.); (N.D.-R.)
| | - Luis Rodríguez-Esparragoza
- Stroke Unit, Department of Neurology, Hospital Germans Trias i Pujol, 08916 Badalona, Catalonia, Spain; (L.R.-E.); (A.D.)
| | - Antoni Dávalos
- Stroke Unit, Department of Neurology, Hospital Germans Trias i Pujol, 08916 Badalona, Catalonia, Spain; (L.R.-E.); (A.D.)
| | - Octavi Martí-Sistac
- Cellular and Molecular Neurobiology Research Group, Department of Neurosciences, Germans Trias i Pujol Research Institute, 08916 Badalona, Catalonia, Spain; (M.M.-S.); (N.D.-R.)
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08916 Bellaterra, Catalonia, Spain
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Carretera del Canyet, Camí de les Escoles s/n, Edifici Mar, 08916 Badalona, Catalonia, Spain
- Correspondence: (O.M.-S.); (T.G.); Tel.: +34-930330531 (O.M.-S.)
| | - Teresa Gasull
- Cellular and Molecular Neurobiology Research Group, Department of Neurosciences, Germans Trias i Pujol Research Institute, 08916 Badalona, Catalonia, Spain; (M.M.-S.); (N.D.-R.)
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Carretera del Canyet, Camí de les Escoles s/n, Edifici Mar, 08916 Badalona, Catalonia, Spain
- Correspondence: (O.M.-S.); (T.G.); Tel.: +34-930330531 (O.M.-S.)
| |
Collapse
|
47
|
Hartnell D, Schwehr BJ, Gillespie-Jones K, Alwis D, Rajan R, Hou H, Sylvain NJ, Pushie MJ, Kelly ME, Massi M, Hackett MJ. Imaging lipophilic regions in rodent brain tissue with halogenated BODIPY probes. Analyst 2020; 145:3809-3813. [PMID: 32400812 DOI: 10.1039/d0an00099j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The effect of halogen substitution in fluorescent BODIPY species was evaluated in the context of staining lipids in situ within brain tissue sections. Herein we demonstrate that the halogenated species maintain their known in vitro affinity when applied to detect lipids in situ in brain tissue sections. Interestingly, the chlorine substituted compound revealed the highest specificify for white matter lipids. Furthermore, the halogen substituted compounds rapidly detected lipid enriched cells, in situ, associated with a case of brain pathology and neuroinflammation.
Collapse
Affiliation(s)
- David Hartnell
- School of Molecular and Life Sciences, Curtin Institute of Functional Molecules and Interfaces, Curtin University, Perth 6845, WA, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Rapid and efficient saltatory action potential conduction depends on the myelin sheath and clustered Na+ channels at nodes of Ranvier. A new study convincingly shows that the periaxonal space is a necessary conductive component to accurately model myelinated axon physiology and saltatory conduction.
Collapse
Affiliation(s)
- Brian C Lim
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
49
|
Pericyte-Mediated Tissue Repair through PDGFRβ Promotes Peri-Infarct Astrogliosis, Oligodendrogenesis, and Functional Recovery after Acute Ischemic Stroke. eNeuro 2020; 7:ENEURO.0474-19.2020. [PMID: 32046974 PMCID: PMC7070447 DOI: 10.1523/eneuro.0474-19.2020] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 01/15/2020] [Accepted: 01/23/2020] [Indexed: 12/16/2022] Open
Abstract
Post-stroke functional recovery can occur spontaneously during the subacute phase; however, how post-stroke fibrotic repair affects functional recovery is highly debated. Platelet-derived growth factor receptor β (PDGFRβ)-expressing pericytes are responsible for post-stroke fibrotic repair within infarct areas; therefore, we examined peri-infarct neural reorganization and functional recovery after permanent middle cerebral artery occlusion (pMCAO) using pericyte-deficient Pdgfrb+/- mice. Time-dependent reduction of infarct area sizes, i.e., repair, was significantly impaired in Pdgfrb+/- mice with recovery of cerebral blood flow (CBF) in ischemic areas attenuated by defective leptomeningeal arteriogenesis and intrainfarct angiogenesis. Peri-infarct astrogliosis, accompanied by increased STAT3 phosphorylation, was attenuated in Pdgfrb+/- mice. Pericyte-conditioned medium (PCM), particularly when treated with platelet-derived growth factor subunit B (PDGFB) homodimer (PDGF-BB; PCM/PDGF-BB), activated STAT3 and enhanced the proliferation and activity of cultured astrocytes. Although peri-infarct proliferation of oligodendrocyte (OL) precursor cells (OPCs) was induced promptly after pMCAO regardless of intrainfarct repair, OPC differentiation and remyelination were significantly attenuated in Pdgfrb+/- mice. Consistently, astrocyte-CM (ACM) promoted OPC differentiation and myelination, which were enhanced remarkably by adding PCM/PDGF-BB to the medium. Post-stroke functional recovery correlated well with the extent and process of intrainfarct repair and peri-infarct oligodendrogenesis. Overall, pericyte-mediated intrainfarct fibrotic repair through PDGFRβ may promote functional recovery through enhancement of peri-infarct oligodendrogenesis as well as astrogliosis after acute ischemic stroke.
Collapse
|
50
|
Xu S, Lu J, Shao A, Zhang JH, Zhang J. Glial Cells: Role of the Immune Response in Ischemic Stroke. Front Immunol 2020; 11:294. [PMID: 32174916 PMCID: PMC7055422 DOI: 10.3389/fimmu.2020.00294] [Citation(s) in RCA: 377] [Impact Index Per Article: 75.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 02/05/2020] [Indexed: 12/16/2022] Open
Abstract
Ischemic stroke, which accounts for 75-80% of all strokes, is the predominant cause of morbidity and mortality worldwide. The post-stroke immune response has recently emerged as a new breakthrough target in the treatment strategy for ischemic stroke. Glial cells, including microglia, astrocytes, and oligodendrocytes, are the primary components of the peri-infarct environment in the central nervous system (CNS) and have been implicated in post-stroke immune regulation. However, increasing evidence suggests that glial cells exert beneficial and detrimental effects during ischemic stroke. Microglia, which survey CNS homeostasis and regulate innate immune responses, are rapidly activated after ischemic stroke. Activated microglia release inflammatory cytokines that induce neuronal tissue injury. By contrast, anti-inflammatory cytokines and neurotrophic factors secreted by alternatively activated microglia are beneficial for recovery after ischemic stroke. Astrocyte activation and reactive gliosis in ischemic stroke contribute to limiting brain injury and re-establishing CNS homeostasis. However, glial scarring hinders neuronal reconnection and extension. Neuroinflammation affects the demyelination and remyelination of oligodendrocytes. Myelin-associated antigens released from oligodendrocytes activate peripheral T cells, thereby resulting in the autoimmune response. Oligodendrocyte precursor cells, which can differentiate into oligodendrocytes, follow an ischemic stroke and may result in functional recovery. Herein, we discuss the mechanisms of post-stroke immune regulation mediated by glial cells and the interaction between glial cells and neurons. In addition, we describe the potential roles of various glial cells at different stages of ischemic stroke and discuss future intervention targets.
Collapse
Affiliation(s)
- Shenbin Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianan Lu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - John H Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, United States.,Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, CA, United States.,Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Brain Research Institute, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, China
| |
Collapse
|