1
|
Zhang YX, Li LY, Xing Y, Chen AX, Xie ST, Li HZ, Zhang QP, Zhang XY, Yang X, Yung WH, Zhu JN. Glutamatergic synaptic plasticity in medial vestibular nuclei during vestibular compensation. Neuroscience 2025:S0306-4522(25)00342-2. [PMID: 40316005 DOI: 10.1016/j.neuroscience.2025.04.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/27/2025] [Accepted: 04/28/2025] [Indexed: 05/04/2025]
Abstract
Vestibular compensation, the spontaneous recovery from vestibular dysfunction following unilateral vestibular loss, serves as a valuable model for investigating post-lesion plasticity in the adult central nervous system. Elucidating the mechanisms underlying vestibular compensation also offers promising therapeutic avenues for treating vestibular disorders. While most studies have focused on the dynamics of GABAergic synaptic plasticity and intrinsic cellular adaptations in the ipsilesional medial vestibular nucleus (MVN) after unilateral labyrinthectomy (UL), the role of glutamatergic synaptic plasticity in this process remains largely unexplored. Here, we employed Golgi staining, immunofluorescence, whole-cell patch-clamp recordings, and behavioral assessments to examine the structural and functional dynamics of glutamatergic synapses during vestibular compensation. Our results reveal rapid structural and functional plasticity of glutamatergic transmission in response to UL. Specifically, dendritic spine density and morphology in the ipsilesional MVN recovered to baseline levels within 6 to 24 h post-UL. Furthermore, UL-induced postsynaptic depression of glutamatergic synaptic strength, reflected by a reduced AMPA/NMDA ratio, was reversed within 24 h, likely due to an upregulation of Ca2+-permeable AMPA receptors. In contrast, presynaptic glutamate release probability, as indicated by a reduced frequency of spontaneous excitatory postsynaptic currents, was not fully compensated during this period. These results suggest that while presynaptic properties recover more slowly in ipsilesional MVN neurons following UL, postsynaptic glutamatergic transmission undergoes rapid structural and functional reorganization. The findings highlight glutamatergic synaptic plasticity as a critical driver for vestibular compensation and suggest that pharmacological interventions targeting these mechanisms may accelerate functional recovery, offering potential therapeutic avenues for vestibular disorders.
Collapse
Affiliation(s)
- Yang-Xun Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Lu-Yao Li
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yue Xing
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Ao-Xue Chen
- Department of Neurology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, China
| | - Shu-Tao Xie
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Hong-Zhao Li
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Qi-Peng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China; Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Xiao-Yang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China; Institute for Brain Sciences, Nanjing University, Nanjing, China.
| | - Xu Yang
- Department of Neurology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, China.
| | - Wing-Ho Yung
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China.
| | - Jing-Ning Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China; Institute for Brain Sciences, Nanjing University, Nanjing, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, China.
| |
Collapse
|
2
|
Raudales A, Schager B, Hancock D, Narayana K, Sharma S, Reeson P, Oshanyk A, Cheema M, Körbelin J, Brown CE. Angiogenesis in the mature mouse cortex is governed in a regional- and Notch1-dependent manner. Cell Rep 2024; 43:115029. [PMID: 39612246 DOI: 10.1016/j.celrep.2024.115029] [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: 05/31/2024] [Revised: 10/10/2024] [Accepted: 11/13/2024] [Indexed: 12/01/2024] Open
Abstract
Cerebral angiogenesis is well appreciated in development and after injury, but the extent to which it occurs across cortical regions in normal adult mice and the underlying mechanisms are incompletely understood. Using in vivo imaging, we show that angiogenesis in anterior-medial cortical regions (retrosplenial and sensorimotor cortex) was exceptionally rare. By contrast, angiogenesis was significantly elevated in posterior-lateral regions such as visual cortex, primarily within 200 μm of the cortical surface. There was no effect of sex on angiogenesis rates, nor were there regional differences in vessel pruning (for either sex). To understand the mechanisms, we surveyed gene expression and found that Notch-related genes were enriched in ultra-stable retrosplenial cortex. Using endothelial-specific knockdown of Notch1, cerebral angiogenesis was significantly increased along with genes implicated in angiogenesis (Apln, Angpt2, Cdkn1a). Our study shows that angiogenesis is regionally dependent and that manipulations of Notch1 could unlock the angiogenic potential of the mature vasculature.
Collapse
Affiliation(s)
- Alejandra Raudales
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Ben Schager
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Dominique Hancock
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Kamal Narayana
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Sorabh Sharma
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Patrick Reeson
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Adam Oshanyk
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Manjinder Cheema
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Jakob Körbelin
- Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Craig E Brown
- Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada; Department of Psychiatry, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| |
Collapse
|
3
|
Goh AR, Park J, Sim AY, Koo BN, Lee YH, Kim JY, Lee JE. Modulating monocyte-derived macrophage polarization in cerebral ischemic injury with hyperglycemia. Exp Neurol 2024; 378:114824. [PMID: 38777250 DOI: 10.1016/j.expneurol.2024.114824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/08/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024]
Abstract
Ischemic stroke (IS), characterized by high mortality rate, occurs owing to diminished or blocked blood flow to the brain. Hyperglycemia (HG) is a major contributor to the risk of IS. HG induces augmented oxidative stress and Blood-Brain Barrier breakdown, which increases the influx of blood-derived myeloid cells into the brain parenchyma. In cerebral ischemia, infiltrating monocytes undergo differentiation into pro-inflammatory or anti-inflammatory macrophages, having a large effect on outcomes of ischemic stroke. In addition, interleukin-4 (IL-4) and interleukin-13 (IL-13) engage in post-ischemia repair by polarizing the infiltrating monocytes into an anti-inflammatory phenotype. In this study, we aimed to determine the effect of phenotypic polarization of monocyte-derived macrophages on the prognosis of IS with HG (HG-IS). We first established a hyperglycemic mouse model using streptozotocin (150 mg/kg) and induced transient middle cerebral artery occlusion. We observed that blood-brain barrier permeability increased in HG-IS mice, as per two-photon live imaging and Evans blue staining. We also confirmed the increased infiltration of monocyte-derived macrophages and the downregulation of anti-inflammatory macrophages related to tissue remodeling after inflammation in HG-IS mice through immunohistochemistry, western blotting, and flow cytometry. We observed phenotypic changes in monocyte-derived macrophages, alleviated infarct volume, and improved motor function in HG-IS mice treated with IL-4 and IL-13. These findings suggest that the modulation of phenotypic changes in monocyte-derived macrophages following IS in hyperglycemic mice may influence ischemic recovery.
Collapse
Affiliation(s)
- A Ra Goh
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea; Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joohyun Park
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - A Young Sim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea; Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Bon-Nyeo Koo
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Ho Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Systems Biology, Glycosylation Network Research Center, Yonsei University, Seoul, Republic of Korea; Interdisciplinary Program of Integrated OMICS for Biomedical Science, Yonsei University, Seoul, Republic of Korea
| | - Jong Youl Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea; Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Yu J, Joo IL, Bazzigaluppi P, Koletar MM, Cherin E, Stanisz AG, Graham JWC, Demore C, Stefanovic B. Micro-ultrasound based characterization of cerebrovasculature following focal ischemic stroke and upon short-term rehabilitation. J Cereb Blood Flow Metab 2024; 44:461-476. [PMID: 37974304 PMCID: PMC10981404 DOI: 10.1177/0271678x231215004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/21/2023] [Accepted: 10/27/2023] [Indexed: 11/19/2023]
Abstract
Notwithstanding recanalization treatments in the acute stage of stroke, many survivors suffer long-term impairments. Physical rehabilitation is the only widely available strategy for chronic-stage recovery, but its optimization is hindered by limited understanding of its effects on brain structure and function. Using micro-ultrasound, behavioral testing, and electrophysiology, we investigated the impact of skilled reaching rehabilitation on cerebral hemodynamics, motor function, and neuronal activity in a rat model of focal ischemic stroke. A 50 MHz micro-ultrasound transducer and intracortical electrophysiology were utilized to characterize neurovascular changes three weeks following focal ischemia elicited by endothelin-1 injection into the sensorimotor cortex. Sprague-Dawley rats were rehabilitated through tray reaching, and their fine skilled reaching was assessed via the Montoya staircase. Focal ischemia led to a sustained deficit in forelimb reaching; and increased tortuosity of the penetrating vessels in the perilesional cortex; with no lateralization of spontaneous neuronal activity. Rehabilitation improved skilled reaching; decreased cortical vascularity; was associated with elevated peri- vs. contralesional hypercapnia-induced flow homogenization and increased perilesional spontaneous cortical neuronal activity. Our study demonstrated neurovascular plasticity accompanying rehabilitation-elicited functional recovery in the subacute stage following stroke, and multiple micro-ultrasound-based markers of cerebrovascular structure and function modified in recovery from ischemia and upon rehabilitation.
Collapse
Affiliation(s)
- Johnson Yu
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Illsung L Joo
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Paolo Bazzigaluppi
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- MetaCell, Cagliari, Italy
| | - Margaret M Koletar
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Emmanuel Cherin
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Andrew G Stanisz
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - James WC Graham
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Christine Demore
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Bojana Stefanovic
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Boyle BR, Berghella AP, Blanco-Suarez E. Astrocyte Regulation of Neuronal Function and Survival in Stroke Pathophysiology. ADVANCES IN NEUROBIOLOGY 2024; 39:233-267. [PMID: 39190078 DOI: 10.1007/978-3-031-64839-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
The interactions between astrocytes and neurons in the context of stroke play crucial roles in the disease's progression and eventual outcomes. After a stroke, astrocytes undergo significant changes in their morphology, molecular profile, and function, together termed reactive astrogliosis. Many of these changes modulate how astrocytes relate to neurons, inducing mechanisms both beneficial and detrimental to stroke recovery. For example, excessive glutamate release and astrocytic malfunction contribute to excitotoxicity in stroke, eventually causing neuronal death. Astrocytes also provide essential metabolic support and neurotrophic signals to neurons after stroke, ensuring homeostatic stability and promoting neuronal survival. Furthermore, several astrocyte-secreted molecules regulate synaptic plasticity in response to stroke, allowing for the rewiring of neural circuits to compensate for damaged areas. In this chapter, we highlight the current understanding of the interactions between astrocytes and neurons in response to stroke, explaining the varied mechanisms contributing to injury progression and the potential implications for future therapeutic interventions.
Collapse
Affiliation(s)
- Bridget R Boyle
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrea P Berghella
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Elena Blanco-Suarez
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Neurological Surgery, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Campos B, Choi H, DeMarco AT, Seydell-Greenwald A, Hussain SJ, Joy MT, Turkeltaub PE, Zeiger W. Rethinking Remapping: Circuit Mechanisms of Recovery after Stroke. J Neurosci 2023; 43:7489-7500. [PMID: 37940595 PMCID: PMC10634578 DOI: 10.1523/jneurosci.1425-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/21/2023] [Accepted: 08/21/2023] [Indexed: 11/10/2023] Open
Abstract
Stroke is one of the most common causes of disability, and there are few treatments that can improve recovery after stroke. Therapeutic development has been hindered because of a lack of understanding of precisely how neural circuits are affected by stroke, and how these circuits change to mediate recovery. Indeed, some of the hypotheses for how the CNS changes to mediate recovery, including remapping, redundancy, and diaschisis, date to more than a century ago. Recent technological advances have enabled the interrogation of neural circuits with ever greater temporal and spatial resolution. These techniques are increasingly being applied across animal models of stroke and to human stroke survivors, and are shedding light on the molecular, structural, and functional changes that neural circuits undergo after stroke. Here we review these studies and highlight important mechanisms that underlie impairment and recovery after stroke. We begin by summarizing knowledge about changes in neural activity that occur in the peri-infarct cortex, specifically considering evidence for the functional remapping hypothesis of recovery. Next, we describe the importance of neural population dynamics, disruptions in these dynamics after stroke, and how allocation of neurons into spared circuits can restore functionality. On a more global scale, we then discuss how effects on long-range pathways, including interhemispheric interactions and corticospinal tract transmission, contribute to post-stroke impairments. Finally, we look forward and consider how a deeper understanding of neural circuit mechanisms of recovery may lead to novel treatments to reduce disability and improve recovery after stroke.
Collapse
Affiliation(s)
- Baruc Campos
- Department of Neurology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California 90095
| | - Hoseok Choi
- Department of Neurology, Weill Institute for Neuroscience, University of California-San Francisco, San Francisco, California 94158
| | - Andrew T DeMarco
- Center for Brain Plasticity and Recovery, Georgetown University Medical Center, Georgetown University, Washington, DC 20057
- Department of Rehabilitation Medicine, Georgetown University Medical Center, Georgetown University, Washington, DC 20057
| | - Anna Seydell-Greenwald
- Center for Brain Plasticity and Recovery, Georgetown University Medical Center, Georgetown University, Washington, DC 20057
- MedStar National Rehabilitation Hospital, Washington, DC 20010
| | - Sara J Hussain
- Movement and Cognitive Rehabilitation Science Program, Department of Kinesiology and Health Education, University of Texas at Austin, Austin, Texas 78712
| | - Mary T Joy
- The Jackson Laboratory, Bar Harbor, Maine 04609
| | - Peter E Turkeltaub
- Center for Brain Plasticity and Recovery, Georgetown University Medical Center, Georgetown University, Washington, DC 20057
- MedStar National Rehabilitation Hospital, Washington, DC 20010
| | - William Zeiger
- Department of Neurology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California 90095
| |
Collapse
|
7
|
Williamson MR, Le SP, Franzen RL, Donlan NA, Rosow JL, Nicot-Cartsonis MS, Cervantes A, Deneen B, Dunn AK, Jones TA, Drew MR. Subventricular zone cytogenesis provides trophic support for neural repair in a mouse model of stroke. Nat Commun 2023; 14:6341. [PMID: 37816732 PMCID: PMC10564905 DOI: 10.1038/s41467-023-42138-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023] Open
Abstract
Stroke enhances proliferation of neural precursor cells within the subventricular zone (SVZ) and induces ectopic migration of newborn cells towards the site of injury. Here, we characterize the identity of cells arising from the SVZ after stroke and uncover a mechanism through which they facilitate neural repair and functional recovery. With genetic lineage tracing, we show that SVZ-derived cells that migrate towards cortical photothrombotic stroke in mice are predominantly undifferentiated precursors. We find that ablation of neural precursor cells or conditional knockout of VEGF impairs neuronal and vascular reparative responses and worsens recovery. Replacement of VEGF is sufficient to induce neural repair and recovery. We also provide evidence that CXCL12 from peri-infarct vasculature signals to CXCR4-expressing cells arising from the SVZ to direct their ectopic migration. These results support a model in which vasculature surrounding the site of injury attracts cells from the SVZ, and these cells subsequently provide trophic support that drives neural repair and recovery.
Collapse
Affiliation(s)
- Michael R Williamson
- Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA.
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA.
| | - Stephanie P Le
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Ronald L Franzen
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
- School of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Nicole A Donlan
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Jill L Rosow
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| | | | - Alexis Cervantes
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience and Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Benjamin Deneen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
- Center for Cancer Neuroscience and Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Andrew K Dunn
- Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Theresa A Jones
- Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Michael R Drew
- Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
- Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
8
|
Wu QL, Cui LY, Ma WY, Wang SS, Zhang Z, Feng ZP, Sun HS, Chu SF, He WB, Chen NH. A novel small-molecular CCR5 antagonist promotes neural repair after stroke. Acta Pharmacol Sin 2023; 44:1935-1947. [PMID: 37198412 PMCID: PMC10545720 DOI: 10.1038/s41401-023-01100-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 04/25/2023] [Indexed: 05/19/2023]
Abstract
Chemokine receptor 5 (CCR5) is one of the main co-receptors of HIV-1, and has been found to be a potential therapeutic target for stroke. Maraviroc is a classic CCR5 antagonist, which is undergoing clinical trials against stroke. As maraviroc shows poor blood-brain barrier (BBB) permeability, it is of interest to find novel CCR5 antagonists suitable for neurological medication. In this study we characterized the therapeutic potential of a novel CCR5 antagonist A14 in treating ischemic stroke mice. A14 was discovered in screening millions compounds in the Chemdiv library based on the molecular docking diagram of CCR5 and maraviroc. We found that A14 dose-dependently inhibited the CCR5 activity with an IC50 value of 4.29 μM. Pharmacodynamic studies showed that A14 treatment exerted protective effects against neuronal ischemic injury both in vitro and vivo. In a SH-SY5Y cell line overexpressing CCR5, A14 (0.1, 1 μM) significantly alleviated OGD/R-induced cell injury. We found that the expression of CCR5 and its ligand CKLF1 was significantly upregulated during both acute and recovery period in focal cortical stroke mice; oral administration of A14 (20 mg·kg-1·d-1, for 1 week) produced sustained protective effect against motor impairment. A14 treatment had earlier onset time, lower onset dosage and much better BBB permeability compared to maraviroc. MRI analysis also showed that A14 treatment significantly reduced the infarction volume after 1 week of treatment. We further revealed that A14 treatment blocked the protein-protein interaction between CCR5 and CKLF1, increasing the activity of CREB signaling pathway in neurons, thereby improving axonal sprouting and synaptic density after stroke. In addition, A14 treatment remarkably inhibited the reactive proliferation of glial cells after stroke and reduced the infiltration of peripheral immune cells. These results demonstrate that A14 is a promising novel CCR5 antagonist for promoting neuronal repair after ischemic stroke. A14 blocked the protein-protein interaction between CKLF1 and CCR5 after stroke by binding with CCR5 stably, improved the infarct area and promoted motor recovery through reversing the CREB/pCREB signaling which was inhibited by activated CCR5 Gαi pathway, and benefited to the dendritic spines and axons sprouting.
Collapse
Affiliation(s)
- Qing-Lin Wu
- Clinical Pharmacology Institute, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Li-Yuan Cui
- 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
| | - Wen-Yu Ma
- Clinical Pharmacology Institute, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Sha-Sha Wang
- Clinical Pharmacology Institute, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhao Zhang
- 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
| | - Zhong-Ping Feng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - 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.
| | - Wen-Bin He
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Shanxi University of Chinese Medicine, Taiyuan, 030024, China.
- National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030024, China.
| | - Nai-Hong Chen
- Clinical Pharmacology Institute, 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
|
9
|
Collyer E, Blanco-Suarez E. Astrocytes in stroke-induced neurodegeneration: a timeline. FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1240862. [PMID: 39086680 PMCID: PMC11285566 DOI: 10.3389/fmmed.2023.1240862] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/22/2023] [Indexed: 08/02/2024]
Abstract
Stroke is a condition characterized by sudden deprivation of blood flow to a brain region and defined by different post-injury phases, which involve various molecular and cellular cascades. At an early stage during the acute phase, fast initial cell death occurs, followed by inflammation and scarring. This is followed by a sub-acute or recovery phase when endogenous plasticity mechanisms may promote spontaneous recovery, depending on various factors that are yet to be completely understood. At later time points, stroke leads to greater neurodegeneration compared to healthy controls in both clinical and preclinical studies, this is evident during the chronic phase when recovery slows down and neurodegenerative signatures appear. Astrocytes have been studied in the context of ischemic stroke due to their role in glutamate re-uptake, as components of the neurovascular unit, as building blocks of the glial scar, and synaptic plasticity regulators. All these roles render astrocytes interesting, yet understudied players in the context of stroke-induced neurodegeneration. With this review, we provide a summary of previous research, highlight astrocytes as potential therapeutic targets, and formulate questions about the role of astrocytes in the mechanisms during the acute, sub-acute, and chronic post-stroke phases that may lead to neurorestoration or neurodegeneration.
Collapse
Affiliation(s)
| | - Elena Blanco-Suarez
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
10
|
Li J, Wu X, Fu Y, Nie H, Tang Z. Two-photon microscopy: application advantages and latest progress for in vivo imaging of neurons and blood vessels after ischemic stroke. Rev Neurosci 2023; 34:559-572. [PMID: 36719181 DOI: 10.1515/revneuro-2022-0127] [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: 10/15/2022] [Accepted: 01/02/2023] [Indexed: 02/01/2023]
Abstract
Two-photon microscopy (TPM) plays an important role in the study of the changes of the two important components of neurovascular units (NVU) - neurons and blood vessels after ischemic stroke (IS). IS refers to sudden neurological dysfunction caused by focal cerebral ischemia, which is one of the leading causes of death and disability worldwide. TPM is a new and rapidly developing high-resolution real-time imaging technique used in vivo that has attracted increasing attention from scientists in the neuroscience field. Neurons and blood vessels are important components of neurovascular units, and they undergo great changes after IS to respond to and compensate for ischemic injury. Here, we introduce the characteristics and pre-imaging preparations of TPM, and review the common methods and latest progress of TPM in the neuronal and vascular research for injury and recovery of IS in recent years. With the review, we clearly recognized that the most important advantage of TPM in the study of ischemic stroke is the ability to perform chronic longitudinal imaging of different tissues at a high resolution in vivo. Finally, we discuss the limitations of TPM and the technological advances in recent years.
Collapse
Affiliation(s)
- Jiarui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, P. R. China
| | - Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, P. R. China
| | - Yu Fu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, P. R. China
| | - Hao Nie
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, P. R. China
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, P. R. China
| |
Collapse
|
11
|
Jing K, Chen F, Shi X, Guo J, Liu X. Dual effect of C-C motif chemokine receptor 5 on ischemic stroke: More harm than benefit? Eur J Pharmacol 2023:175857. [PMID: 37321471 DOI: 10.1016/j.ejphar.2023.175857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/02/2023] [Accepted: 06/13/2023] [Indexed: 06/17/2023]
Abstract
Ischemic stroke involves a series of complex pathological mechanisms, of which neuroinflammation is currently the most widely recognized. C-C motif chemokine receptor 5 (CCR5) has recently been shown to be upregulated after cerebral ischemia. Notably, CCR5 is not only involved in neuroinflammation, but also in the blood-brain barrier, neural structures, and connections. Accumulating experimental studies indicate that CCR5 has a dual effect on ischemic stroke. In the acute phase after cerebral ischemia, the pro-inflammatory and disruptive effect of CCR5 on the blood-brain barrier predominates. However, in the chronic phase, the effect of CCR5 on the repair of neural structures and connections is thought to be cell-type dependent. Interestingly, clinical evidence has shown that CCR5 might be harmful rather than beneficial. CCR5-Δ32 mutation or CCR5 antagonist exerts a neuroprotective effect in patients with ischemic stroke. Considering CCR5 as an attractive potential target, we introduce the current research progress of the entangled relationships between CCR5 and ischemic stroke. Clinical data are still needed to determine the efficacy of activating or inactivating CCR5 in the treatment of ischemic stroke, especially for potential phase- or cell type-dependent treatments in the future.
Collapse
Affiliation(s)
- Kai Jing
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Feng Chen
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Xiaofei Shi
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Jinmin Guo
- Department of Clinical Pharmacy, 960th Hospital of Joint Logistic Support Force, Shandong, Jinan, China.
| | - Xia Liu
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China.
| |
Collapse
|
12
|
Xiong H, Tang F, Guo Y, Xu R, Lei P. Neural Circuit Changes in Neurological Disorders: Evidence from in vivo Two-photon Imaging. Ageing Res Rev 2023; 87:101933. [PMID: 37061201 DOI: 10.1016/j.arr.2023.101933] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 04/11/2023] [Indexed: 04/17/2023]
Abstract
Neural circuits, such as synaptic plasticity and neural activity, are critical components of healthy brain function. The consequent dynamic remodeling of neural circuits is an ongoing procedure affecting neuronal activities. Disruption of this essential process results in diseases. Advanced microscopic applications such as two-photon laser scanning microscopy have recently been applied to understand neural circuit changes during disease since it can visualize fine structural and functional cellular activation in living animals. In this review, we have summarized the latest work assessing the dynamic rewiring of postsynaptic dendritic spines and modulation of calcium transients in neurons of the intact living brain, focusing on their potential roles in neurological disorders (e.g. Alzheimer's disease, stroke, and epilepsy). Understanding the fine changes that occurred in the brain during disease is crucial for future clinical intervention developments.
Collapse
Affiliation(s)
- Huan Xiong
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China; Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Fei Tang
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Yujie Guo
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, Chengdu, 610041, China.
| |
Collapse
|
13
|
Qiao C, Liu Z, Qie S. The Implications of Microglial Regulation in Neuroplasticity-Dependent Stroke Recovery. Biomolecules 2023; 13:biom13030571. [PMID: 36979506 PMCID: PMC10046452 DOI: 10.3390/biom13030571] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/23/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Stroke causes varying degrees of neurological deficits, leading to corresponding dysfunctions. There are different therapeutic principles for each stage of pathological development. Neuroprotection is the main treatment in the acute phase, and functional recovery becomes primary in the subacute and chronic phases. Neuroplasticity is considered the basis of functional restoration and neurological rehabilitation after stroke, including the remodeling of dendrites and dendritic spines, axonal sprouting, myelin regeneration, synapse shaping, and neurogenesis. Spatiotemporal development affects the spontaneous rewiring of neural circuits and brain networks. Microglia are resident immune cells in the brain that contribute to homeostasis under physiological conditions. Microglia are activated immediately after stroke, and phenotypic polarization changes and phagocytic function are crucial for regulating focal and global brain inflammation and neurological recovery. We have previously shown that the development of neuroplasticity is spatiotemporally consistent with microglial activation, suggesting that microglia may have a profound impact on neuroplasticity after stroke and may be a key therapeutic target for post-stroke rehabilitation. In this review, we explore the impact of neuroplasticity on post-stroke restoration as well as the functions and mechanisms of microglial activation, polarization, and phagocytosis. This is followed by a summary of microglia-targeted rehabilitative interventions that influence neuroplasticity and promote stroke recovery.
Collapse
Affiliation(s)
- Chenye Qiao
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Zongjian Liu
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Shuyan Qie
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| |
Collapse
|
14
|
Ding J, Dai Y, Zhu J, Fan X, Zhang H, Tang B. Research advances in cGAS-stimulator of interferon genes pathway and central nervous system diseases: Focus on new therapeutic approaches. Front Mol Neurosci 2022; 15:1050837. [PMID: 36618820 PMCID: PMC9817143 DOI: 10.3389/fnmol.2022.1050837] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Cyclic GMP-AMP synthase (cGAS), a crucial innate immune sensor, recognizes cytosolic DNA and induces stimulator of interferon genes (STING) to produce type I interferon and other proinflammatory cytokines, thereby mediating innate immune signaling. The cGAS-STING pathway is involved in the regulation of infectious diseases, anti-tumor immunity, and autoimmune diseases; in addition, it plays a key role in the development of central nervous system (CNS) diseases. Therapeutics targeting the modulation of cGAS-STING have promising clinical applications. Here, we summarize the cGAS-STING signaling mechanism and the recent research on its role in CNS diseases.
Collapse
Affiliation(s)
- Jiao Ding
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yijie Dai
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiahui Zhu
- Department of Neurology, The Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuemei Fan
- Department of Neurology, The Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Zhang
- Department of Neurology, The Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,*Correspondence: Hao Zhang,
| | - Bo Tang
- Department of Neurology, The Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,Bo Tang,
| |
Collapse
|
15
|
Bechay KR, Abduljawad N, Latifi S, Suzuki K, Iwashita H, Carmichael ST. PDE2A Inhibition Enhances Axonal Sprouting, Functional Connectivity, and Recovery after Stroke. J Neurosci 2022; 42:8225-8236. [PMID: 36163142 PMCID: PMC9653274 DOI: 10.1523/jneurosci.0730-22.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 09/06/2022] [Accepted: 09/11/2022] [Indexed: 11/21/2022] Open
Abstract
Phosphodiesterase (PDE) inhibitors have been safely and effectively used in the clinic and increase the concentration of intracellular cyclic nucleotides (cAMP/cGMP). These molecules activate downstream mediators, including the cAMP response element-binding protein (CREB), which controls neuronal excitability and growth responses. CREB gain of function enhances learning and allocates neurons into memory engrams. CREB also controls recovery after stroke. PDE inhibitors are linked to recovery from neural damage and to stroke recovery in specific sites within the brain. PDE2A is enriched in cortex. In the present study, we use a mouse cortical stroke model in young adult and aged male mice to test the effect of PDE2A inhibition on functional recovery, and on downstream mechanisms of axonal sprouting, tissue repair, and the functional connectivity of neurons in recovering cortex. Stroke causes deficits in use of the contralateral forelimb, loss of axonal projections in cortex adjacent to the infarct, and functional disconnection of neuronal networks. PDE2A inhibition enhances functional recovery, increases axonal projections in peri-infarct cortex, and, through two-photon in vivo imaging, enhances the functional connectivity of motor system excitatory neurons. PDE2A inhibition after stroke does not have an effect on other aspects of tissue repair, such as angiogenesis, gliogenesis, neurogenesis, and inflammatory responses. These data suggest that PDE2A inhibition is an effective therapeutic approach for stroke recovery in the rodent and that it simultaneously enhances connectivity in peri-infarct neuronal populations.SIGNIFICANCE STATEMENT Inhibition of PDE2A enhances motor recovery, axonal projections, and functional connectivity of neurons in peri-infarct tissue. This represents an avenue for a pharmacological therapy for stroke recovery.
Collapse
Affiliation(s)
- Kirollos Raouf Bechay
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Nora Abduljawad
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Shahrzad Latifi
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Kazunori Suzuki
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa 251-8555, Japan
| | - Hiroki Iwashita
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa 251-8555, Japan
| | - S Thomas Carmichael
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| |
Collapse
|
16
|
Michiels L, Mertens N, Thijs L, Radwan A, Sunaert S, Vandenbulcke M, Verheyden G, Koole M, Van Laere K, Lemmens R. Changes in synaptic density in the subacute phase after ischemic stroke: A 11C-UCB-J PET/MR study. J Cereb Blood Flow Metab 2022; 42:303-314. [PMID: 34550834 PMCID: PMC9122519 DOI: 10.1177/0271678x211047759] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Functional alterations after ischemic stroke have been described with Magnetic Resonance Imaging (MRI) and perfusion Positron Emission Tomography (PET), but no data on in vivo synaptic changes exist. Recently, imaging of synaptic density became available by targeting synaptic vesicle protein 2 A, a protein ubiquitously expressed in all presynaptic nerve terminals. We hypothesized that in subacute ischemic stroke loss of synaptic density can be evaluated with 11C-UCB-J PET in the ischemic tissue and that alterations in synaptic density can be present in brain regions beyond the ischemic core. We recruited ischemic stroke patients to undergo 11C-UCB-J PET/MR imaging 21 ± 8 days after stroke onset to investigate regional 11C-UCB-J SUVR (standardized uptake value ratio). There was a decrease (but residual signal) of 11C-UCB-J SUVR within the lesion of 16 stroke patients compared to 40 healthy controls (ratiolesion/controls = 0.67 ± 0.28, p = 0.00023). Moreover, 11C-UCB-J SUVR was lower in the non-lesioned tissue of the affected hemisphere compared to the unaffected hemisphere (ΔSUVR = -0.17, p = 0.0035). The contralesional cerebellar hemisphere showed a lower 11C-UCB-J SUVR compared to the ipsilesional cerebellar hemisphere (ΔSUVR = -0.14, p = 0.0048). In 8 out of 16 patients, the asymmetry index suggested crossed cerebellar diaschisis. Future research is required to longitudinally study these changes in synaptic density and their association with outcome.
Collapse
Affiliation(s)
- Laura Michiels
- Department of Neurosciences, KU Leuven, Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Nathalie Mertens
- Nuclear Medicine and Molecular Imaging, 26657KU Leuven, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Liselot Thijs
- Department of Rehabilitation Sciences, 26657KU Leuven, KU Leuven, Leuven, Belgium
| | - Ahmed Radwan
- Translational MRI, 26657KU Leuven, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Stefan Sunaert
- Translational MRI, 26657KU Leuven, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.,Department of Geriatric Psychiatry, University Psychiatric Centre, KU Leuven, Leuven, Belgium
| | - Geert Verheyden
- Department of Rehabilitation Sciences, 26657KU Leuven, KU Leuven, Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, 26657KU Leuven, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, 26657KU Leuven, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,Division of Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Robin Lemmens
- Department of Neurosciences, KU Leuven, Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
17
|
Wu X, Zeng H, Cai L, Chen G. Role of the Extracellular Traps in Central Nervous System. Front Immunol 2021; 12:783882. [PMID: 34868063 PMCID: PMC8635093 DOI: 10.3389/fimmu.2021.783882] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/26/2021] [Indexed: 12/20/2022] Open
Abstract
It has been reported that several immune cells can release chromatin and granular proteins into extracellular space in response to the stimulation, forming extracellular traps (ETs). The cells involved in the extracellular trap formation are recognized including neutropils, macrophages, basophils, eosinophils, and mast cells. With the development of research related to central nervous system, the role of ETs has been valued in neuroinflammation, blood–brain barrier, and other fields. Meanwhile, it has been found that microglial cells as the resident immune cells of the central nervous system can also release ETs, updating the original understanding. This review aims to clarify the role of the ETs in the central nervous system, especially in neuroinflammation and blood–brain barrier.
Collapse
Affiliation(s)
- Xinyan Wu
- Department of Neurological Surgery The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hanhai Zeng
- Department of Neurological Surgery The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lingxin Cai
- Department of Neurological Surgery The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Gao Chen
- Department of Neurological Surgery The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
18
|
Longitudinal functional imaging of VIP interneurons reveals sup-population specific effects of stroke that are rescued with chemogenetic therapy. Nat Commun 2021; 12:6112. [PMID: 34671051 PMCID: PMC8528851 DOI: 10.1038/s41467-021-26405-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 10/04/2021] [Indexed: 01/20/2023] Open
Abstract
Stroke profoundly disrupts cortical excitability which impedes recovery, but how it affects the function of specific inhibitory interneurons, or subpopulations therein, is poorly understood. Interneurons expressing vasoactive intestinal peptide (VIP) represent an intriguing stroke target because they can regulate cortical excitability through disinhibition. Here we chemogenetically augmented VIP interneuron excitability in a murine model of photothrombotic stroke and show that it enhances somatosensory responses and improves recovery of paw function. Using longitudinal calcium imaging, we discovered that stroke primarily disrupts the fidelity (fraction of responsive trials) and predictability of sensory responses within a subset of highly active VIP neurons. Partial recovery of responses occurred largely within these active neurons and was not accompanied by the recruitment of minimally active neurons. Importantly, chemogenetic stimulation preserved sensory response fidelity and predictability in highly active neurons. These findings provide a new depth of understanding into how stroke and prospective therapies (chemogenetics), can influence subpopulations of inhibitory interneurons.
Collapse
|
19
|
Zhang Z, Sun GY, Ding S. Glial Cell Line-Derived Neurotrophic Factor and Focal Ischemic Stroke. Neurochem Res 2021; 46:2638-2650. [PMID: 33591443 PMCID: PMC8364922 DOI: 10.1007/s11064-021-03266-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 11/29/2022]
Abstract
Focal ischemic stroke (FIS) is a leading cause of human debilitation and death. Following the onset of a FIS, the brain experiences a series of spatiotemporal changes which are exemplified in different pathological processes. One prominent feature of FIS is the development of reactive astrogliosis and glial scar formation in the peri-infarct region (PIR). During the subacute phase, astrocytes in PIR are activated, referred to as reactive astrocytes (RAs), exhibit changes in morphology (hypotrophy), show an increased proliferation capacity, and altered gene expression profile, a phenomenon known as reactive astrogliosis. Subsequently, the morphology of RAs remains stable, and proliferation starts to decline together with the formation of glial scars. Reactive astrogliosis and glial scar formation eventually cause substantial tissue remodeling and changes in permanent structure around the PIR. Glial cell line-derived neurotrophic factor (GDNF) was originally isolated from a rat glioma cell-line and regarded as a potent survival neurotrophic factor. Under normal conditions, GDNF is expressed in neurons but is upregulated in RAs after FIS. This review briefly describes properties of GDNF, its receptor-mediated signaling pathways, as well as recent studies regarding the role of RAs-derived GDNF in neuronal protection and brain recovery. These results provide evidence suggesting an important role of RA-derived GDNF in intrinsic brain repair and recovery after FIS, and thus targeting GDNF in RAs may be effective for stroke therapy.
Collapse
Affiliation(s)
- Zhe Zhang
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO, 65211, USA
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri-Columbia, Columbia, MO, 65211, USA
| | - Grace Y Sun
- Department of Biochemistry, University of Missouri-Columbia, Columbia, MO, 65211, USA
| | - Shinghua Ding
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO, 65211, USA.
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri-Columbia, Columbia, MO, 65211, USA.
- Dalton Cardiovascular Research Center, Department of Biomedical, Biological and Chemical Engineering, University of Missouri-Columbia, 134 Research Park Drive, Columbia, MO, 65211, USA.
| |
Collapse
|
20
|
Park J, Kim JY, Kim YR, Huang M, Chang JY, Sim AY, Jung H, Lee WT, Hyun YM, Lee JE. Reparative System Arising from CCR2(+) Monocyte Conversion Attenuates Neuroinflammation Following Ischemic Stroke. Transl Stroke Res 2021; 12:879-893. [PMID: 33409730 PMCID: PMC8421302 DOI: 10.1007/s12975-020-00878-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/20/2020] [Accepted: 11/16/2020] [Indexed: 01/22/2023]
Abstract
Monocytes recruitment from the blood to inflamed tissues following ischemic stroke is an important immune response to wound healing and tissue repair. Mouse monocytes can be endogenously divided into two distinct populations: pro-inflammatory or classical monocytes that express CCR2highCX3CR1low and circulate in blood, and anti-inflammatory or non-classical monocytes that express CCR2lowCX3CR1high and patrol locally. In this study of transgenic mice with functional CX3CR1GFP/+ or CX3CR1GFP/+-CCR2RFP/+, we found that CCR2highCX3CR1low monocytes recruited to the injured brain were cytokine-dependently converted into CCR2lowCX3CR1high macrophages, especially under the influence of IL-4 and IL-13, thereby attenuating the neuroinflammation following sterile ischemic stroke. The overall data suggest that (1) the regulation of monocyte-switching is one of the ultimate reparative strategies in ischemic stroke, and (2) the adaptation of monocytes in a locally inflamed milieu is vital to alleviating the effects of ischemic stroke through innate immunity.
Collapse
Affiliation(s)
- Joohyun Park
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Youl Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yu Rim Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Meiying Huang
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji Young Chang
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - A Young Sim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hosung Jung
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Won Taek Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young-Min Hyun
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
21
|
Tene O, Hallevi H, Molad J, Usher S, Seyman E, Bornstein NM, Shenhar-Tsarfaty S, Ben Assayag E. CCR5-Δ32 polymorphism: a possible protective factor for post-stroke depressive symptoms. J Psychiatry Neurosci 2021; 46:E431-E440. [PMID: 34291627 PMCID: PMC8519488 DOI: 10.1503/jpn.200197] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND A naturally occurring loss-of-function mutation in the gene for C-C chemokine receptor type 5 (CCR5-Δ32) has recently been reported as a protective factor in post-stroke motor and cognitive recovery. We sought to examine whether this mutation also prevented the development of depressive symptoms up to 2 years after a stroke. METHODS Participants were survivors of a first-ever mild to moderate ischemic stroke or transient ischemic attack from the TABASCO prospective study who underwent a 3 T MRI at baseline and were examined by a multiprofessional team 6, 12 and 24 months after the event, including an evaluation of depressive symptoms using the Geriatric Depression Scale. RESULTS CCR5-Δ32 status and a baseline depression evaluation were available for 435 patients. Compared with noncarriers, CCR5-Δ32 carriers (16.1%) had fewer depressive symptoms at admission (p = 0.035) and at 6 months (p < 0.001), 12 months (p < 0.001) and 24 months (p = 0.006) after the index event. This association remained significant at 6 and 12 months after adjustment for age, sex, education, antidepressant use, ethnicity and the presence of cortical infarcts. These findings were more robust in women. Compared to baseline, depressive symptoms in CCR5-Δ32 noncarriers tended to remain stable or grow worse over time, but in CCR5-Δ32 carriers, symptoms tended to improve. LIMITATIONS A limitation of this study was the exclusion of patients who had a severe stroke or who had pre-stroke depression. CONCLUSION Carriers of the CCR5-Δ32 allele had a lower tendency to develop depressive symptoms post-stroke, and this phenomenon was more prominent in women. These findings could have clinical implications; they suggest a mechanism-based treatment target for post-stroke depression. Drugs mimicking this loss-of-function mutation exist and could serve as a novel antidepressant therapy.
Collapse
Affiliation(s)
- Oren Tene
- From the Departments of Neurology and Psychiatry, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel (Tene, Hallevi, Molad, Usher, Seyman, Shenhar-Tsarfaty, Ben Assayag); the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel (Tene, Hallevi, Bornstein, Shenhar-Tsarfaty, Ben Assayag); and the Brain Center, Shaare Zedek Medical Center, Jerusalem, Israel (Bornstein)
| | - Hen Hallevi
- From the Departments of Neurology and Psychiatry, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel (Tene, Hallevi, Molad, Usher, Seyman, Shenhar-Tsarfaty, Ben Assayag); the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel (Tene, Hallevi, Bornstein, Shenhar-Tsarfaty, Ben Assayag); and the Brain Center, Shaare Zedek Medical Center, Jerusalem, Israel (Bornstein)
| | - Jeremy Molad
- From the Departments of Neurology and Psychiatry, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel (Tene, Hallevi, Molad, Usher, Seyman, Shenhar-Tsarfaty, Ben Assayag); the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel (Tene, Hallevi, Bornstein, Shenhar-Tsarfaty, Ben Assayag); and the Brain Center, Shaare Zedek Medical Center, Jerusalem, Israel (Bornstein)
| | - Saly Usher
- From the Departments of Neurology and Psychiatry, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel (Tene, Hallevi, Molad, Usher, Seyman, Shenhar-Tsarfaty, Ben Assayag); the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel (Tene, Hallevi, Bornstein, Shenhar-Tsarfaty, Ben Assayag); and the Brain Center, Shaare Zedek Medical Center, Jerusalem, Israel (Bornstein)
| | - Estelle Seyman
- From the Departments of Neurology and Psychiatry, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel (Tene, Hallevi, Molad, Usher, Seyman, Shenhar-Tsarfaty, Ben Assayag); the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel (Tene, Hallevi, Bornstein, Shenhar-Tsarfaty, Ben Assayag); and the Brain Center, Shaare Zedek Medical Center, Jerusalem, Israel (Bornstein)
| | - Natan M Bornstein
- From the Departments of Neurology and Psychiatry, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel (Tene, Hallevi, Molad, Usher, Seyman, Shenhar-Tsarfaty, Ben Assayag); the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel (Tene, Hallevi, Bornstein, Shenhar-Tsarfaty, Ben Assayag); and the Brain Center, Shaare Zedek Medical Center, Jerusalem, Israel (Bornstein)
| | - Shani Shenhar-Tsarfaty
- From the Departments of Neurology and Psychiatry, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel (Tene, Hallevi, Molad, Usher, Seyman, Shenhar-Tsarfaty, Ben Assayag); the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel (Tene, Hallevi, Bornstein, Shenhar-Tsarfaty, Ben Assayag); and the Brain Center, Shaare Zedek Medical Center, Jerusalem, Israel (Bornstein)
| | - Einor Ben Assayag
- From the Departments of Neurology and Psychiatry, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel (Tene, Hallevi, Molad, Usher, Seyman, Shenhar-Tsarfaty, Ben Assayag); the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel (Tene, Hallevi, Bornstein, Shenhar-Tsarfaty, Ben Assayag); and the Brain Center, Shaare Zedek Medical Center, Jerusalem, Israel (Bornstein)
| |
Collapse
|
22
|
Mirza Agha B, Akbary R, Ghasroddashti A, Nazari-Ahangarkolaee M, Whishaw IQ, Mohajerani MH. Cholinergic upregulation by optogenetic stimulation of nucleus basalis after photothrombotic stroke in forelimb somatosensory cortex improves endpoint and motor but not sensory control of skilled reaching in mice. J Cereb Blood Flow Metab 2021; 41:1608-1622. [PMID: 33103935 PMCID: PMC8221755 DOI: 10.1177/0271678x20968930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A network of cholinergic neurons in the basal forebrain innerve the forebrain and are proposed to contribute to a variety of functions including cortical plasticity, attention, and sensorimotor behavior. This study examined the contribution of the nucleus basalis cholinergic projection to the sensorimotor cortex on recovery on a skilled reach-to-eat task following photothrombotic stroke in the forelimb region of the somatosensory cortex. Mice were trained to perform a single pellet skilled reaching task and their pre and poststroke performance, from Day 4 to Day 28 poststroke, was assessed frame-by-frame by video analysis with endpoint, movement and sensorimotor integration measures. Somatosensory forelimb lesions produced impairments in endpoint and movement component measures of reaching and increased the incidence of fictive eating, a sensory impairment in mistaking a missed reach for a successful reach. Upregulated acetylcholine (ACh) release, as measured by local field potential recording, elicited via optogenetic stimulation of the nucleus basalis improved recovery of reaching and improved movement scores but did not affect sensorimotor integration impairment poststroke. The results show that the mouse cortical forelimb somatosensory region contributes to forelimb motor behavior and suggest that ACh upregulation could serve as an adjunct to behavioral therapy for acute treatment of stroke.
Collapse
Affiliation(s)
- Behroo Mirza Agha
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Roya Akbary
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Arashk Ghasroddashti
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Mojtaba Nazari-Ahangarkolaee
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Ian Q Whishaw
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Majid H Mohajerani
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| |
Collapse
|
23
|
Zeiger WA, Marosi M, Saggi S, Noble N, Samad I, Portera-Cailliau C. Barrel cortex plasticity after photothrombotic stroke involves potentiating responses of pre-existing circuits but not functional remapping to new circuits. Nat Commun 2021; 12:3972. [PMID: 34172735 PMCID: PMC8233353 DOI: 10.1038/s41467-021-24211-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 06/01/2021] [Indexed: 01/14/2023] Open
Abstract
Recovery after stroke is thought to be mediated by adaptive circuit plasticity, whereby surviving neurons assume the roles of those that died. However, definitive longitudinal evidence of neurons changing their response selectivity after stroke is lacking. We sought to directly test whether such functional “remapping” occurs within mouse primary somatosensory cortex after a stroke that destroys the C1 barrel. Using in vivo calcium imaging to longitudinally record sensory-evoked activity under light anesthesia, we did not find any increase in the number of C1 whisker-responsive neurons in the adjacent, spared D3 barrel after stroke. To promote plasticity after stroke, we also plucked all whiskers except C1 (forced use therapy). This led to an increase in the reliability of sensory-evoked responses in C1 whisker-responsive neurons but did not increase the number of C1 whisker-responsive neurons in spared surround barrels over baseline levels. Our results argue against remapping of functionality after barrel cortex stroke, but support a circuit-based mechanism for how rehabilitation may improve recovery. Definitive evidence for functional remapping after stroke remains lacking. Here, the authors performed in vivo intrinsic signal imaging and two-photon calcium imaging of sensory-evoked responses before and after photothrombotic stroke and found no evidence of remapping of lost functionalities to new circuits in peri-infarct cortex.
Collapse
Affiliation(s)
- William A Zeiger
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| | - Máté Marosi
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Satvir Saggi
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Natalie Noble
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Isa Samad
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Carlos Portera-Cailliau
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA. .,Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
24
|
Williamson MR, Fuertes CJA, Dunn AK, Drew MR, Jones TA. Reactive astrocytes facilitate vascular repair and remodeling after stroke. Cell Rep 2021; 35:109048. [PMID: 33910014 PMCID: PMC8142687 DOI: 10.1016/j.celrep.2021.109048] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/30/2021] [Accepted: 04/07/2021] [Indexed: 12/18/2022] Open
Abstract
Brain injury causes astrocytes to assume a reactive state that is essential for early tissue protection, but how reactive astrocytes affect later reparative processes is incompletely understood. In this study, we show that reactive astrocytes are crucial for vascular repair and remodeling after ischemic stroke in mice. Analysis of astrocytic gene expression data reveals substantial activation of transcriptional programs related to vascular remodeling after stroke. In vivo two-photon imaging provides evidence of astrocytes contacting newly formed vessels in cortex surrounding photothrombotic infarcts. Chemogenetic ablation of a subset of reactive astrocytes after stroke dramatically impairs vascular and extracellular matrix remodeling. This disruption of vascular repair is accompanied by prolonged blood flow deficits, exacerbated vascular permeability, ongoing cell death, and worsened motor recovery. In contrast, vascular structure in the non-ischemic brain is unaffected by focal astrocyte ablation. These findings position reactive astrocytes as critical cellular mediators of functionally important vascular remodeling during neural repair.
Collapse
Affiliation(s)
- Michael R Williamson
- Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA.
| | | | - Andrew K Dunn
- Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA; Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Michael R Drew
- Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA; Center for Learning and Memory, University of Texas at Austin, Austin, TX 78712, USA; Department of Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| | - Theresa A Jones
- Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA; Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
25
|
Abstract
Stroke is a debilitating disease. Current effective therapies for stroke recovery are limited to neurorehabilitation. Most stroke recovery occurs in a limited and early time window. Many of the mechanisms of spontaneous recovery after stroke parallel mechanisms of normal learning and memory. While various efforts are in place to identify potential drug targets, an emerging approach is to understand biological correlates between learning and stroke recovery. This review assesses parallels between biological changes at the molecular, structural, and functional levels during learning and recovery after stroke, with a focus on drug and cellular targets for therapeutics.
Collapse
Affiliation(s)
- Mary Teena Joy
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - S. Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
26
|
Joy MT, Carmichael ST. Encouraging an excitable brain state: mechanisms of brain repair in stroke. Nat Rev Neurosci 2021; 22:38-53. [PMID: 33184469 PMCID: PMC10625167 DOI: 10.1038/s41583-020-00396-7] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2020] [Indexed: 02/02/2023]
Abstract
Stroke induces a plastic state in the brain. This period of enhanced plasticity leads to the sprouting of new axons, the formation of new synapses and the remapping of sensory-motor functions, and is associated with motor recovery. This is a remarkable process in the adult brain, which is normally constrained in its levels of neuronal plasticity and connectional change. Recent evidence indicates that these changes are driven by molecular systems that underlie learning and memory, such as changes in cellular excitability during memory formation. This Review examines circuit changes after stroke, the shared mechanisms between memory formation and brain repair, the changes in neuronal excitability that underlie stroke recovery, and the molecular and pharmacological interventions that follow from these findings to promote motor recovery in animal models. From these findings, a framework emerges for understanding recovery after stroke, central to which is the concept of neuronal allocation to damaged circuits. The translation of the concepts discussed here to recovery in humans is underway in clinical trials for stroke recovery drugs.
Collapse
Affiliation(s)
- Mary T Joy
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
27
|
Sunil S, Evren Erdener S, Cheng X, Kura S, Tang J, Jiang J, Karrobi K, Kılıç K, Roblyer D, Boas DA. Stroke core revealed by tissue scattering using spatial frequency domain imaging. NEUROIMAGE-CLINICAL 2020; 29:102539. [PMID: 33385882 PMCID: PMC7779322 DOI: 10.1016/j.nicl.2020.102539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/26/2022]
Abstract
We present OCT and SFDI as methods to measure the spatial extent of stroke in mice. OCT was a reliable predictor of the stroke core in a photothrombosis stroke model. SFDI scattering coefficient spatially overlaps with OCT attenuation after stroke. Scattering increases following stroke reliably predict ischemic injury. SFDI provides a wide-field optical approach to map the stroke core.
Ischemic stroke leads to a reduction or complete loss of blood supply causing injury to brain tissue, which ultimately leads to behavioral impairment. Optical techniques are widely used to study the structural and functional changes that result as a consequence of ischemic stroke both in the acute and chronic phases of stroke recovery. It is currently a challenge to accurately estimate the spatial extent of the infarct without the use of histological parameters however, and in order to follow recovery mechanisms longitudinally at the mesoscopic scale it is essential to know the spatial extent of the stroke core. In this paper we first establish optical coherence tomography (OCT) as a reliable indicator of the stroke core by analyzing signal attenuation and spatially correlating it with the infarct, determined by staining with triphenyl-tetrazolium chloride (TTC). We then introduce spatial frequency domain imaging (SFDI) as a mesoscopic optical technique that can be used to accurately measure the infarct spatial extent by exploiting changes in optical scattering that occur as a consequence of ischemic stroke. Additionally, we follow the progression of ischemia through the acute and sub-acute phases of stroke recovery using both OCT and SFDI and show a consistently high spatial overlap in estimating infarct location. The use of SFDI in assessing infarct location will allow longitudinal studies targeted at following functional recovery mechanisms on a mesoscopic level without having to sacrifice the mouse acutely.
Collapse
Affiliation(s)
- Smrithi Sunil
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| | - Sefik Evren Erdener
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Xiaojun Cheng
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Sreekanth Kura
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Jianbo Tang
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - John Jiang
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Kavon Karrobi
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Kıvılcım Kılıç
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Darren Roblyer
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Department of Electrical and Computer Engineering, Boston University, Boston, MA 02215, USA
| | - David A Boas
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Department of Electrical and Computer Engineering, Boston University, Boston, MA 02215, USA
| |
Collapse
|
28
|
Schager B, Brown CE. Susceptibility to capillary plugging can predict brain region specific vessel loss with aging. J Cereb Blood Flow Metab 2020; 40:2475-2490. [PMID: 31903837 PMCID: PMC7820682 DOI: 10.1177/0271678x19895245] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/28/2019] [Accepted: 11/19/2019] [Indexed: 12/13/2022]
Abstract
Vessel loss in the aging brain is commonly reported, yet important questions remain concerning whether there are regional vulnerabilities and what mechanisms could account for these regional differences, if they exist. Here we imaged and quantified vessel length, tortuosity and width in 15 brain regions in young adult and aged mice. Our data indicate that vessel loss was most pronounced in white matter followed by cortical, then subcortical grey matter regions, while some regions (visual cortex, amygdala, thalamus) showed no decline with aging. Regions supplied by the anterior cerebral artery were more vulnerable to loss than those supplied by middle or posterior cerebral arteries. Vessel width and tortuosity generally increased with age but neither reliably predicted regional vessel loss. Since capillaries are naturally prone to plugging and prolonged obstructions often lead to vessel pruning, we hypothesized that regional susceptibilities to plugging could help predict vessel loss. By mapping the distribution of microsphere-induced capillary obstructions, we discovered that regions with a higher density of persistent obstructions were more likely to show vessel loss with aging and vice versa. These findings indicate that age-related vessel loss is region specific and can be explained, at least partially, by regional susceptibilities to capillary plugging.
Collapse
Affiliation(s)
- Ben Schager
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Craig E Brown
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
29
|
Allegra Mascaro AL, Conti E, Lai S, Di Giovanna AP, Spalletti C, Alia C, Panarese A, Scaglione A, Sacconi L, Micera S, Caleo M, Pavone FS. Combined Rehabilitation Promotes the Recovery of Structural and Functional Features of Healthy Neuronal Networks after Stroke. Cell Rep 2020; 28:3474-3485.e6. [PMID: 31553915 DOI: 10.1016/j.celrep.2019.08.062] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 06/19/2019] [Accepted: 08/20/2019] [Indexed: 10/26/2022] Open
Abstract
Rehabilitation is considered the most effective treatment for promoting the recovery of motor deficits after stroke. One of the most challenging experimental goals is to unambiguously link brain rewiring to motor improvement prompted by rehabilitative therapy. Previous work showed that robotic training combined with transient inactivation of the contralesional cortex promotes a generalized recovery in a mouse model of stroke. Here, we use advanced optical imaging and manipulation tools to study cortical remodeling induced by this rehabilitation paradigm. We show that the stabilization of peri-infarct synaptic contacts accompanies increased vascular density induced by angiogenesis. Furthermore, temporal and spatial features of cortical activation recover toward pre-stroke conditions through the progressive formation of a new motor representation in the peri-infarct area. In the same animals, we observe reinforcement of inter-hemispheric connectivity. Our results provide evidence that combined rehabilitation promotes the restoration of structural and functional features distinctive of healthy neuronal networks.
Collapse
Affiliation(s)
- Anna Letizia Allegra Mascaro
- Neuroscience Institute, National Research Council, Pisa 56124, Italy; European Laboratory for Non-Linear Spectroscopy, University of Florence, Sesto Fiorentino 50019, Italy.
| | - Emilia Conti
- European Laboratory for Non-Linear Spectroscopy, University of Florence, Sesto Fiorentino 50019, Italy; Department of Physics and Astronomy, University of Florence, Sesto Fiorentino 50019, Italy
| | - Stefano Lai
- Translational Neural Engineering Area, The BioRobotics Institute, Scuola Superiore Sant'Anna, Pisa 56127, Italy
| | | | | | - Claudia Alia
- Neuroscience Institute, National Research Council, Pisa 56124, Italy
| | - Alessandro Panarese
- Translational Neural Engineering Area, The BioRobotics Institute, Scuola Superiore Sant'Anna, Pisa 56127, Italy
| | - Alessandro Scaglione
- European Laboratory for Non-Linear Spectroscopy, University of Florence, Sesto Fiorentino 50019, Italy
| | - Leonardo Sacconi
- European Laboratory for Non-Linear Spectroscopy, University of Florence, Sesto Fiorentino 50019, Italy; National Institute of Optics, National Research Council, Sesto Fiorentino 50019, Italy
| | - Silvestro Micera
- Translational Neural Engineering Area, The BioRobotics Institute, Scuola Superiore Sant'Anna, Pisa 56127, Italy; Bertarelli Foundation Chair in Translational NeuroEngineering, Centre for Neuroprosthetics and Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Matteo Caleo
- Neuroscience Institute, National Research Council, Pisa 56124, Italy; Department of Biomedical Sciences, University of Padua, Padova 35131, Italy
| | - Francesco Saverio Pavone
- European Laboratory for Non-Linear Spectroscopy, University of Florence, Sesto Fiorentino 50019, Italy; Department of Physics and Astronomy, University of Florence, Sesto Fiorentino 50019, Italy; National Institute of Optics, National Research Council, Sesto Fiorentino 50019, Italy
| |
Collapse
|
30
|
Williamson MR, Franzen RL, Fuertes CJA, Dunn AK, Drew MR, Jones TA. A Window of Vascular Plasticity Coupled to Behavioral Recovery after Stroke. J Neurosci 2020; 40:7651-7667. [PMID: 32873722 PMCID: PMC7531554 DOI: 10.1523/jneurosci.1464-20.2020] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/03/2020] [Accepted: 08/25/2020] [Indexed: 12/13/2022] Open
Abstract
Stroke causes remodeling of vasculature surrounding the infarct, but whether and how vascular remodeling contributes to recovery are unclear. We established an approach to monitor and compare changes in vascular structure and blood flow with high spatiotemporal precision after photothrombotic infarcts in motor cortex using longitudinal 2-photon and multiexposure speckle imaging in mice of both sexes. A spatially graded pattern of vascular structural remodeling in peri-infarct cortex unfolded over the first 2 weeks after stroke, characterized by vessel loss and formation, and selective stabilization of a subset of new vessels. This vascular structural plasticity was coincident with transient activation of transcriptional programs relevant for vascular remodeling, reestablishment of peri-infarct blood flow, and large improvements in motor performance. Local vascular plasticity was strongly predictive of restoration of blood flow, which was in turn predictive of behavioral recovery. These findings reveal the spatiotemporal evolution of vascular remodeling after stroke and demonstrate that a window of heightened vascular plasticity is coupled to the reestablishment of blood flow and behavioral recovery. Our findings support that neovascularization contributes to behavioral recovery after stroke by restoring blood flow to peri-infarct regions. These findings may inform strategies for enhancing recovery from stroke and other types of brain injury.SIGNIFICANCE STATEMENT An improved understanding of neural repair could inform strategies for enhancing recovery from stroke and other types of brain injury. Stroke causes remodeling of vasculature surrounding the lesion, but whether and how the process of vascular remodeling contributes to recovery of behavioral function have been unclear. Here we used longitudinal in vivo imaging to track vascular structure and blood flow in residual peri-infarct cortex after ischemic stroke in mice. We found that stroke created a restricted period of heightened vascular plasticity that was associated with restoration of blood flow, which was in turn predictive of recovery of motor function. Therefore, our findings support that vascular remodeling facilitates behavioral recovery after stroke by restoring blood flow to peri-infarct cortex.
Collapse
Affiliation(s)
| | | | | | - Andrew K Dunn
- Institute for Neuroscience
- Department of Biomedical Engineering
| | - Michael R Drew
- Institute for Neuroscience
- Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, Texas 78712
| | | |
Collapse
|
31
|
Freitas-Andrade M, Raman-Nair J, Lacoste B. Structural and Functional Remodeling of the Brain Vasculature Following Stroke. Front Physiol 2020; 11:948. [PMID: 32848875 PMCID: PMC7433746 DOI: 10.3389/fphys.2020.00948] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Maintenance of cerebral blood vessel integrity and regulation of cerebral blood flow ensure proper brain function. The adult human brain represents only a small portion of the body mass, yet about a quarter of the cardiac output is dedicated to energy consumption by brain cells at rest. Due to a low capacity to store energy, brain health is heavily reliant on a steady supply of oxygen and nutrients from the bloodstream, and is thus particularly vulnerable to stroke. Stroke is a leading cause of disability and mortality worldwide. By transiently or permanently limiting tissue perfusion, stroke alters vascular integrity and function, compromising brain homeostasis and leading to widespread consequences from early-onset motor deficits to long-term cognitive decline. While numerous lines of investigation have been undertaken to develop new pharmacological therapies for stroke, only few advances have been made and most clinical trials have failed. Overall, our understanding of the acute and chronic vascular responses to stroke is insufficient, yet a better comprehension of cerebrovascular remodeling following stroke is an essential prerequisite for developing novel therapeutic options. In this review, we present a comprehensive update on post-stroke cerebrovascular remodeling, an important and growing field in neuroscience, by discussing cellular and molecular mechanisms involved, sex differences, limitations of preclinical research design and future directions.
Collapse
Affiliation(s)
| | - Joanna Raman-Nair
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
32
|
Zhang N, Zhang Z, He R, Li H, Ding S. GLAST-CreER T2 mediated deletion of GDNF increases brain damage and exacerbates long-term stroke outcomes after focal ischemic stroke in mouse model. Glia 2020; 68:2395-2414. [PMID: 32497340 DOI: 10.1002/glia.23848] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 01/01/2023]
Abstract
Focal ischemic stroke (FIS) is a leading cause of human death. Glial scar formation largely caused by reactive astrogliosis in peri-infarct region (PIR) is the hallmark of FIS. Glial cell-derived neurotrophic factor (GDNF) was originally isolated from a rat glioma cell-line supernatant and is a potent survival neurotrophic factor. Here, using CreERT2 -LoxP recombination technology, we generated inducible and astrocyte-specific GDNF conditional knockout (cKO), that is, GLAST-GDNF-/- cKO mice to investigate the effect of reactive astrocytes (RAs)-derived GDNF on neuronal death, brain damage, oxidative stress and motor function recovery after photothrombosis (PT)-induced FIS. Under non-ischemic conditions, we found that adult GLAST-GDNF-/- cKO mice exhibited significant lower numbers of Brdu+, Ki67+ cells, and DCX+ cells in the dentate gyrus (DG) in hippocampus than GDNF floxed (GDNFf/f ) control (Ctrl) mice, indicating endogenous astrocytic GDNF can promote adult neurogenesis. Under ischemic conditions, GLAST-GDNF-/- cKO mice had a significant increase in infarct volume, hippocampal damage and FJB+ degenerating neurons after PT as compared with the Ctrl mice. GLAST-GDNF-/- cKO mice also had lower densities of Brdu+ and Ki67+ cells in the PIR and exhibited larger behavioral deficits than the Ctrl mice. Mechanistically, GDNF deficiency in astrocytes increased oxidative stress through the downregulation of glucose-6-phosphate dehydrogenase (G6PD) in RAs. In summary, our study indicates that RAs-derived endogenous GDNF plays important roles in reducing brain damage and promoting brain recovery after FIS through neural regeneration and suggests that promoting anti-oxidant mechanism in RAs is a potential strategy in stroke therapy.
Collapse
Affiliation(s)
- Nannan Zhang
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Zhe Zhang
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, USA
| | - Rui He
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Hailong Li
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA.,Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, USA
| | - Shinghua Ding
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA.,Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
33
|
Neutrophil extracellular traps released by neutrophils impair revascularization and vascular remodeling after stroke. Nat Commun 2020; 11:2488. [PMID: 32427863 PMCID: PMC7237502 DOI: 10.1038/s41467-020-16191-y] [Citation(s) in RCA: 310] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 04/20/2020] [Indexed: 01/13/2023] Open
Abstract
Neovascularization and vascular remodeling are functionally important for brain repair after stroke. We show that neutrophils accumulate in the peri-infarct cortex during all stages of ischemic stroke. Neutrophils producing intravascular and intraparenchymal neutrophil extracellular traps (NETs) peak at 3-5 days. Neutrophil depletion reduces blood-brain barrier (BBB) breakdown and enhances neovascularization at 14 days. Peptidylarginine deiminase 4 (PAD4), an enzyme essential for NET formation, is upregulated in peri-ischemic brains. Overexpression of PAD4 induces an increase in NET formation that is accompanied by reduced neovascularization and increased BBB damage. Disruption of NETs by DNase 1 and inhibition of NET formation by genetic ablation or pharmacologic inhibition of PAD increases neovascularization and vascular repair and improves functional recovery. Furthermore, PAD inhibition reduces stroke-induced STING-mediated production of IFN-β, and STING knockdown and IFN receptor-neutralizing antibody treatment reduces BBB breakdown and increases vascular plasticity. Collectively, our results indicate that NET release impairs vascular remodeling during stroke recovery.
Collapse
|
34
|
He F, Sullender CT, Zhu H, Williamson MR, Li X, Zhao Z, Jones TA, Xie C, Dunn AK, Luan L. Multimodal mapping of neural activity and cerebral blood flow reveals long-lasting neurovascular dissociations after small-scale strokes. SCIENCE ADVANCES 2020; 6:eaba1933. [PMID: 32494746 PMCID: PMC7244270 DOI: 10.1126/sciadv.aba1933] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/10/2020] [Indexed: 06/02/2023]
Abstract
Neurovascular coupling, the close spatial and temporal relationship between neural activity and hemodynamics, is disrupted in pathological brain states. To understand the altered neurovascular relationship in brain disorders, longitudinal, simultaneous mapping of neural activity and hemodynamics is critical yet challenging to achieve. Here, we use a multimodal neural platform in a mouse model of stroke and realize long-term, spatially resolved tracking of intracortical neural activity and cerebral blood flow in the same brain regions. We observe a pronounced neurovascular dissociation that occurs immediately after small-scale strokes, becomes the most severe a few days after, lasts into chronic periods, and varies with the level of ischemia. Neuronal deficits extend spatiotemporally, whereas restoration of cerebral blood flow occurs sooner and reaches a higher relative value. Our findings reveal the neurovascular impact of ministrokes and inform the limitation of neuroimaging techniques that infer neural activity from hemodynamic responses.
Collapse
Affiliation(s)
- Fei He
- Department of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
- Neuroengineering Initiative, Rice University, 6500 Main Street, Houston, TX 77005, USA
| | - Colin T. Sullender
- Department of Biomedical Engineering, The University of Texas at Austin, 107 E. Dean Keeton Street, 1 University Station, C0800, Austin, TX 78712, USA
| | - Hanlin Zhu
- Department of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
| | - Michael R. Williamson
- Institute for Neuroscience, The University of Texas at Austin, 1 University Station, Stop C7000, Austin, TX 78712, USA
| | - Xue Li
- Department of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
| | - Zhengtuo Zhao
- Department of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
- Neuroengineering Initiative, Rice University, 6500 Main Street, Houston, TX 77005, USA
| | - Theresa A. Jones
- Department of Psychology, The University of Texas at Austin, 108 E. Dean Keeton Street, Stop A8000, SEA 6.106, Austin, TX 78712, USA
| | - Chong Xie
- Department of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
- Neuroengineering Initiative, Rice University, 6500 Main Street, Houston, TX 77005, USA
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
| | - Andrew K. Dunn
- Department of Biomedical Engineering, The University of Texas at Austin, 107 E. Dean Keeton Street, 1 University Station, C0800, Austin, TX 78712, USA
| | - Lan Luan
- Department of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
- Neuroengineering Initiative, Rice University, 6500 Main Street, Houston, TX 77005, USA
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
| |
Collapse
|
35
|
Joy MT, Ben Assayag E, Shabashov-Stone D, Liraz-Zaltsman S, Mazzitelli J, Arenas M, Abduljawad N, Kliper E, Korczyn AD, Thareja NS, Kesner EL, Zhou M, Huang S, Silva TK, Katz N, Bornstein NM, Silva AJ, Shohami E, Carmichael ST. CCR5 Is a Therapeutic Target for Recovery after Stroke and Traumatic Brain Injury. Cell 2020; 176:1143-1157.e13. [PMID: 30794775 DOI: 10.1016/j.cell.2019.01.044] [Citation(s) in RCA: 253] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 10/05/2018] [Accepted: 01/24/2019] [Indexed: 12/19/2022]
Abstract
We tested a newly described molecular memory system, CCR5 signaling, for its role in recovery after stroke and traumatic brain injury (TBI). CCR5 is uniquely expressed in cortical neurons after stroke. Post-stroke neuronal knockdown of CCR5 in pre-motor cortex leads to early recovery of motor control. Recovery is associated with preservation of dendritic spines, new patterns of cortical projections to contralateral pre-motor cortex, and upregulation of CREB and DLK signaling. Administration of a clinically utilized FDA-approved CCR5 antagonist, devised for HIV treatment, produces similar effects on motor recovery post stroke and cognitive decline post TBI. Finally, in a large clinical cohort of stroke patients, carriers for a naturally occurring loss-of-function mutation in CCR5 (CCR5-Δ32) exhibited greater recovery of neurological impairments and cognitive function. In summary, CCR5 is a translational target for neural repair in stroke and TBI and the first reported gene associated with enhanced recovery in human stroke.
Collapse
Affiliation(s)
- Mary T Joy
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Einor Ben Assayag
- Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dalia Shabashov-Stone
- Department of Pharmacology, The Institute for Drug Research, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sigal Liraz-Zaltsman
- Department of Pharmacology, The Institute for Drug Research, Hebrew University of Jerusalem, Jerusalem, Israel; The Joseph Sagol Neuroscience Center, Sheba Medical Center, Israel; Institute for Health and Medical Professions, Ono Academic College, Kiryat Ono, Israel
| | - Jose Mazzitelli
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Marcela Arenas
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Nora Abduljawad
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Efrat Kliper
- Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Amos D Korczyn
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nikita S Thareja
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Efrat L Kesner
- Department of Pharmacology, The Institute for Drug Research, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Miou Zhou
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory and Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Shan Huang
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory and Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Tawnie K Silva
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory and Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Noomi Katz
- Institute for Health and Medical Professions, Ono Academic College, Kiryat Ono, Israel
| | - Natan M Bornstein
- Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alcino J Silva
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory and Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Esther Shohami
- Department of Pharmacology, The Institute for Drug Research, Hebrew University of Jerusalem, Jerusalem, Israel
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
36
|
Regenhardt RW, Takase H, Lo EH, Lin DJ. Translating concepts of neural repair after stroke: Structural and functional targets for recovery. Restor Neurol Neurosci 2020; 38:67-92. [PMID: 31929129 PMCID: PMC7442117 DOI: 10.3233/rnn-190978] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stroke is among the most common causes of adult disability worldwide, and its disease burden is shifting towards that of a long-term condition. Therefore, the development of approaches to enhance recovery and augment neural repair after stroke will be critical. Recovery after stroke involves complex interrelated systems of neural repair. There are changes in both structure (at the molecular, cellular, and tissue levels) and function (in terms of excitability, cortical maps, and networks) that occur spontaneously within the brain. Several approaches to augment neural repair through enhancing these changes are under study. These include identifying novel drug targets, implementing rehabilitation strategies, and developing new neurotechnologies. Each of these approaches has its own array of different proposed mechanisms. Current investigation has emphasized both cellular and circuit-based targets in both gray and white matter, including axon sprouting, dendritic branching, neurogenesis, axon preservation, remyelination, blood brain barrier integrity, blockade of extracellular inhibitory signals, alteration of excitability, and promotion of new brain cortical maps and networks. Herein, we review for clinicians recovery after stroke, basic elements of spontaneous neural repair, and ongoing work to augment neural repair. Future study requires alignment of basic, translational, and clinical research. The field continues to grow while becoming more clearly defined. As thrombolysis changed stroke care in the 1990 s and thrombectomy in the 2010 s, the augmentation of neural repair and recovery after stroke may revolutionize care for these patients in the coming decade.
Collapse
Affiliation(s)
- Robert W Regenhardt
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| | - Hajime Takase
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| | - Eng H Lo
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| | - David J Lin
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
- Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| |
Collapse
|
37
|
Hayden EY, Putman J, Nunez S, Shin WS, Oberoi M, Charreton M, Dutta S, Li Z, Komuro Y, Joy MT, Bitan G, MacKenzie-Graham A, Jiang L, Hinman JD. Ischemic axonal injury up-regulates MARK4 in cortical neurons and primes tau phosphorylation and aggregation. Acta Neuropathol Commun 2019; 7:135. [PMID: 31429800 PMCID: PMC6700776 DOI: 10.1186/s40478-019-0783-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 08/01/2019] [Indexed: 12/11/2022] Open
Abstract
Ischemic injury to white matter tracts is increasingly recognized to play a key role in age-related cognitive decline, vascular dementia, and Alzheimer’s disease. Knowledge of the effects of ischemic axonal injury on cortical neurons is limited yet critical to identifying molecular pathways that link neurodegeneration and ischemia. Using a mouse model of subcortical white matter ischemic injury coupled with retrograde neuronal tracing, we employed magnetic affinity cell sorting with fluorescence-activated cell sorting to capture layer-specific cortical neurons and performed RNA-sequencing. With this approach, we identified a role for microtubule reorganization within stroke-injured neurons acting through the regulation of tau. We find that subcortical stroke-injured Layer 5 cortical neurons up-regulate the microtubule affinity-regulating kinase, Mark4, in response to axonal injury. Stroke-induced up-regulation of Mark4 is associated with selective remodeling of the apical dendrite after stroke and the phosphorylation of tau in vivo. In a cell-based tau biosensor assay, Mark4 promotes the aggregation of human tau in vitro. Increased expression of Mark4 after ischemic axonal injury in deep layer cortical neurons provides new evidence for synergism between axonal and neurodegenerative pathologies by priming of tau phosphorylation and aggregation.
Collapse
|
38
|
Durán-Laforet V, Fernández-López D, García-Culebras A, González-Hijón J, Moraga A, Palma-Tortosa S, García-Yébenes I, Vega-Pérez A, Lizasoain I, Moro MÁ. Delayed Effects of Acute Reperfusion on Vascular Remodeling and Late-Phase Functional Recovery After Stroke. Front Neurosci 2019; 13:767. [PMID: 31396042 PMCID: PMC6664024 DOI: 10.3389/fnins.2019.00767] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 07/09/2019] [Indexed: 11/24/2022] Open
Abstract
Tissue perfusion is a necessary condition for vessel survival that can be compromised under ischemic conditions. Following stroke, delayed effects of early brain reperfusion on the vascular substrate necessary for remodeling, perfusion and maintenance of proper peri-lesional hemodynamics are unknown. Such aspects of ischemic injury progression may be critical for neurological recovery in stroke patients. This study aims to describe the impact of early, non-thrombolytic reperfusion on the vascular brain component and its potential contribution to tissue remodeling and long-term functional recovery beyond the acute phase after stroke in 3-month-old male C57bl/6 mice. Permanent (pMCAO) and transient (60 min, tMCAO) brain ischemia mouse models were used for characterizing the effect of early, non-thrombolytic reperfusion on the brain vasculature. Analysis of different vascular parameters (vessel density, proliferation, degeneration and perfusion) revealed that, while early middle cerebral artery recanalization was not sufficient to prevent sub-acute vascular degeneration within the ischemic brain regions, brain reperfusion promoted a secondary wave of vascular remodeling in the peri-lesional regions, which led to improved perfusion of the ischemic boundaries and late-phase neurological recovery. This study concluded that acute, non-thrombolytic artery recanalization following stroke favors late-phase vascular remodeling and improves peri-lesional perfusion, contributing to secondary functional recovery.
Collapse
Affiliation(s)
- Violeta Durán-Laforet
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - David Fernández-López
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Alicia García-Culebras
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Juan González-Hijón
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Ana Moraga
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Sara Palma-Tortosa
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Isaac García-Yébenes
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Adrián Vega-Pérez
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Ignacio Lizasoain
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - María Ángeles Moro
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| |
Collapse
|
39
|
Voglewede RL, Vandemark KM, Davidson AM, DeWitt AR, Heffler MD, Trimmer EH, Mostany R. Reduced sensory-evoked structural plasticity in the aging barrel cortex. Neurobiol Aging 2019; 81:222-233. [PMID: 31323444 DOI: 10.1016/j.neurobiolaging.2019.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 06/15/2019] [Accepted: 06/15/2019] [Indexed: 10/26/2022]
Abstract
Impairments in synaptic connectivity have been linked to cognitive deficits in age-related neurodegenerative disorders and healthy aging. However, the anatomical and structural bases of these impairments have not been identified yet. A hallmark of neural plasticity in young adults is short-term synaptic rearrangement, yet aged animals already display higher synaptic turnover rates at the baseline. Using two-photon excitation (2PE) microscopy, we explored if this elevated turnover alters the aged brain's response to plasticity. Following a sensory-evoked plasticity protocol involving whisker stimulation, aged mice display reduced spine dynamics (gain, loss, and turnover), decreased spine clustering, and lower spine stability when compared to young adult mice. These results suggest a deficiency of the cortical neurons of aged mice to structurally incorporate new sensory experiences, in the form of clustered, long-lasting synapses, into already existing cortical circuits. This research provides the first evidence linking experience-dependent plasticity with in vivo spine dynamics in the aged brain and supports a model of both reduced synaptic plasticity and reduced synaptic tenacity in the aged somatosensory system.
Collapse
Affiliation(s)
- Rebecca L Voglewede
- Neuroscience Program, Tulane University School of Science and Engineering, New Orleans, LA, USA; Tulane Brain Institute, Tulane University, New Orleans, LA, USA; Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Kaeli M Vandemark
- Neuroscience Program, Tulane University School of Science and Engineering, New Orleans, LA, USA; Tulane Brain Institute, Tulane University, New Orleans, LA, USA
| | - Andrew M Davidson
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA; Department of Cell and Molecular Biology, Tulane University School of Science and Engineering, New Orleans, LA, USA
| | - Annie R DeWitt
- Neuroscience Program, Tulane University School of Science and Engineering, New Orleans, LA, USA; Tulane Brain Institute, Tulane University, New Orleans, LA, USA
| | - Marissa D Heffler
- Neuroscience Program, Tulane University School of Science and Engineering, New Orleans, LA, USA; Tulane Brain Institute, Tulane University, New Orleans, LA, USA; Department of Biomedical Engineering, Tulane University School of Science and Engineering, Lindy Boggs Center Suite 500, New Orleans, LA, USA
| | - Emma H Trimmer
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ricardo Mostany
- Neuroscience Program, Tulane University School of Science and Engineering, New Orleans, LA, USA; Tulane Brain Institute, Tulane University, New Orleans, LA, USA; Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
40
|
Sandvig I, Augestad IL, Håberg AK, Sandvig A. Neuroplasticity in stroke recovery. The role of microglia in engaging and modifying synapses and networks. Eur J Neurosci 2018; 47:1414-1428. [PMID: 29786167 DOI: 10.1111/ejn.13959] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/13/2018] [Accepted: 04/17/2018] [Indexed: 02/06/2023]
Abstract
Neuroplasticity after ischaemic injury involves both spontaneous rewiring of neural networks and circuits as well as functional responses in neurogenic niches. These events involve complex interactions with activated microglia, which evolve in a dynamic manner over time. Although the exact mechanisms underlying these interactions remain poorly understood, increasing experimental evidence suggests a determining role of pro- and anti-inflammatory microglial activation profiles in shaping both synaptogenesis and neurogenesis. While the inflammatory response of microglia was thought to be detrimental, a more complex profile of the role of microglia in tissue remodelling is emerging. Experimental evidence suggests that microglia in response to injury can rapidly modify neuronal activity and modulate synaptic function, as well as be beneficial for the proliferation and integration of neural progenitor cells (NPCs) from endogenous neurogenic niches into functional networks thereby supporting stroke recovery. The manner in which microglia contribute towards sculpting neural synapses and networks, both in terms of activity-dependent and homeostatic plasticity, suggests that microglia-mediated pro- and/or anti-inflammatory activity may significantly contribute towards spontaneous neuronal plasticity after ischaemic lesions. In this review, we first introduce some of the key cellular and molecular mechanisms underlying neuroplasticity in stroke and then proceed to discuss the crosstalk between microglia and endogenous neuroplasticity in response to brain ischaemia with special focus on the engagement of synapses and neural networks and their implications for grey matter integrity and function in stroke repair.
Collapse
Affiliation(s)
- Ioanna Sandvig
- Faculty of Medicine and Health Sciences, Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ingrid Lovise Augestad
- Faculty of Medicine and Health Sciences, Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Asta Kristine Håberg
- Faculty of Medicine and Health Sciences, Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Radiology and Nuclear Medicine, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Axel Sandvig
- Faculty of Medicine and Health Sciences, Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Neurology, St Olav's Hospital, Trondheim University Hospital, Trondheim, Norway.,Department of Pharmacology and Clinical Neurosciences, Division of Neuro, Head and Neck, Umeå University Hospital, Umeå, Sweden
| |
Collapse
|
41
|
Reeson P, Choi K, Brown CE. VEGF signaling regulates the fate of obstructed capillaries in mouse cortex. eLife 2018; 7:e33670. [PMID: 29697373 PMCID: PMC5919759 DOI: 10.7554/elife.33670] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/19/2018] [Indexed: 12/12/2022] Open
Abstract
Cortical capillaries are prone to obstruction, which over time, could have a major impact on brain angioarchitecture and function. The mechanisms that govern the removal of these obstructions and what long-term fate awaits obstructed capillaries, remains a mystery. We estimate that ~0.12% of mouse cortical capillaries are obstructed each day (lasting >20 min), preferentially in superficial layers and lower order branches. Tracking natural or microsphere-induced obstructions revealed that 75-80% of capillaries recanalized within 24 hr. Remarkably, 30% of all obstructed capillaries were pruned by 21 days, including some that had regained flow. Pruning involved regression of endothelial cells, which was not compensated for by sprouting. Using this information, we predicted capillary loss with aging that closely matched experimental estimates. Genetic knockdown or inhibition of VEGF-R2 signaling was a critical factor in promoting capillary recanalization and minimizing subsequent pruning. Our studies reveal the incidence, mechanism and long-term outcome of capillary obstructions which can also explain age-related capillary rarefaction.
Collapse
Affiliation(s)
- Patrick Reeson
- Division of Medical SciencesUniversity of VictoriaVictoriaCanada
| | - Kevin Choi
- Division of Medical SciencesUniversity of VictoriaVictoriaCanada
| | - Craig E Brown
- Division of Medical SciencesUniversity of VictoriaVictoriaCanada
- Department of BiologyUniversity of VictoriaVictoriaCanada
- Department of PsychiatryUniversity of British ColumbiaVancouverCanada
| |
Collapse
|
42
|
Alexander BH, Barnes HM, Trimmer E, Davidson AM, Ogola BO, Lindsey SH, Mostany R. Stable Density and Dynamics of Dendritic Spines of Cortical Neurons Across the Estrous Cycle While Expressing Differential Levels of Sensory-Evoked Plasticity. Front Mol Neurosci 2018; 11:83. [PMID: 29615867 PMCID: PMC5864847 DOI: 10.3389/fnmol.2018.00083] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/01/2018] [Indexed: 12/11/2022] Open
Abstract
Periodic oscillations of gonadal hormone levels during the estrous cycle exert effects on the female brain, impacting cognition and behavior. While previous research suggests that changes in hormone levels across the cycle affect dendritic spine dynamics in the hippocampus, little is known about the effects on cortical dendritic spines and previous studies showed contradictory results. In this in vivo imaging study, we investigated the impact of the estrous cycle on the density and dynamics of dendritic spines of pyramidal neurons in the primary somatosensory cortex of mice. We also examined if the induction of synaptic plasticity during proestrus, estrus, and metestrus/diestrus had differential effects on the degree of remodeling of synapses in this brain area. We used chronic two-photon excitation (2PE) microscopy during steady-state conditions and after evoking synaptic plasticity by whisker stimulation at the different stages of the cycle. We imaged apical dendritic tufts of layer 5 pyramidal neurons of naturally cycling virgin young female mice. Spine density, turnover rate (TOR), survival fraction, morphology, and volume of mushroom spines remained unaltered across the estrous cycle, and the values of these parameters were comparable with those of young male mice. However, while whisker stimulation of female mice during proestrus and estrus resulted in increases in the TOR of spines (74.2 ± 14.9% and 75.1 ± 12.7% vs. baseline, respectively), sensory-evoked plasticity was significantly lower during metestrus/diestrus (32.3 ± 12.8%). In males, whisker stimulation produced 46.5 ± 20% increase in TOR compared with baseline—not significantly different from female mice at any stage of the cycle. These results indicate that, while steady-state density and dynamics of dendritic spines of layer 5 pyramidal neurons in the primary somatosensory cortex of female mice are constant during the estrous cycle, the susceptibility of these neurons to sensory-evoked structural plasticity may be dependent on the stage of the cycle. Since dendritic spines are more plastic during proestrus and estrus than during metestrus/diestrus, certain stages of the cycle could be more suitable for forms of memory requiring de novo formation and elimination of spines and other stages for forms of memory where retention and/or repurposing of already existing synaptic connections is more pertinent.
Collapse
Affiliation(s)
- Bailin H Alexander
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States
| | - Heather M Barnes
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States.,Neuroscience Program, Brain Institute, Tulane University, New Orleans, LA, United States
| | - Emma Trimmer
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States
| | - Andrew M Davidson
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States.,Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| | - Benard O Ogola
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States.,Brain Institute, Tulane University, New Orleans, LA, United States
| | - Ricardo Mostany
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States.,Brain Institute, Tulane University, New Orleans, LA, United States
| |
Collapse
|
43
|
Kim SY, Hsu JE, Husbands LC, Kleim JA, Jones TA. Coordinated Plasticity of Synapses and Astrocytes Underlies Practice-Driven Functional Vicariation in Peri-Infarct Motor Cortex. J Neurosci 2018; 38:93-107. [PMID: 29133435 PMCID: PMC5761439 DOI: 10.1523/jneurosci.1295-17.2017] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 10/05/2017] [Accepted: 11/03/2017] [Indexed: 01/05/2023] Open
Abstract
Motor rehabilitative training after stroke can improve motor function and promote topographical reorganization of remaining motor cortical movement representations, but this reorganization follows behavioral improvements. A more detailed understanding of the neural bases of rehabilitation efficacy is needed to inform therapeutic efforts to improve it. Using a rat model of upper extremity impairments after ischemic stroke, we examined effects of motor rehabilitative training at the ultrastructural level in peri-infarct motor cortex. Extensive training in a skilled reaching task promoted improved performance and recovery of more normal movements. This was linked with greater axodendritic synapse density and ultrastructural characteristics of enhanced synaptic efficacy that were coordinated with changes in perisynaptic astrocytic processes in the border region between head and forelimb areas of peri-infarct motor cortex. Disrupting synapses and motor maps by infusions of anisomycin (ANI) into anatomically reorganized motor, but not posterior parietal, cortex eliminated behavioral gains from rehabilitative training. In contrast, ANI infusion in the equivalent cortical region of intact animals had no effect on reaching skills. These results suggest that rehabilitative training efficacy for improving manual skills is mediated by synaptic plasticity in a region of motor cortex that, before lesions, is not essential for manual skills, but becomes so as a result of the training. These findings support that experience-driven synaptic structural reorganization underlies functional vicariation in residual motor cortex after motor cortical infarcts.SIGNIFICANCE STATEMENT Stroke is a leading cause of long-term disability. Motor rehabilitation, the main treatment for physical disability, is of variable efficacy. A better understanding of neural mechanisms underlying effective motor rehabilitation would inform strategies for improving it. Here, we reveal synaptic underpinnings of effective motor rehabilitation. Rehabilitative training improved manual skill in the paretic forelimb and induced the formation of special synapse subtypes in coordination with structural changes in astrocytes, a glial cell that influences neural communication. These changes were found in a region that is nonessential for manual skill in intact animals, but came to mediate this skill due to training after stroke. Therefore, motor rehabilitation efficacy depends on synaptic changes that enable remaining brain regions to assume new functions.
Collapse
Affiliation(s)
- Soo Young Kim
- Department of Integrative Biology, University of California, Berkeley, California 94720,
| | - J Edward Hsu
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, Texas 77030
- Institute for Neuroscience
| | | | - Jeffrey A Kleim
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287
| | - Theresa A Jones
- Institute for Neuroscience
- Psychology Department, University of Texas, Austin, Texas 78712, and
| |
Collapse
|
44
|
Marked bias towards spontaneous synaptic inhibition distinguishes non-adapting from adapting layer 5 pyramidal neurons in the barrel cortex. Sci Rep 2017; 7:14959. [PMID: 29097689 PMCID: PMC5668277 DOI: 10.1038/s41598-017-14971-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 10/19/2017] [Indexed: 11/18/2022] Open
Abstract
Pyramidal neuron subtypes differ in intrinsic electrophysiology properties and dendritic morphology. However, do different pyramidal neuron subtypes also receive synaptic inputs that are dissimilar in frequency and in excitation/inhibition balance? Unsupervised clustering of three intrinsic parameters that vary by cell subtype – the slow afterhyperpolarization, the sag, and the spike frequency adaptation – split layer 5 barrel cortex pyramidal neurons into two clusters: one of adapting cells and one of non-adapting cells, corresponding to previously described thin- and thick-tufted pyramidal neurons, respectively. Non-adapting neurons presented frequencies of spontaneous inhibitory postsynaptic currents (sIPSCs) and spontaneous excitatory postsynaptic currents (sEPSCs) three- and two-fold higher, respectively, than those of adapting neurons. The IPSC difference between pyramidal subtypes was activity independent. A subset of neurons were thy1-GFP positive, presented characteristics of non-adapting pyramidal neurons, and also had higher IPSC and EPSC frequencies than adapting neurons. The sEPSC/sIPSC frequency ratio was higher in adapting than in non-adapting cells, suggesting a higher excitatory drive in adapting neurons. Therefore, our study on spontaneous synaptic inputs suggests a different extent of synaptic information processing in adapting and non-adapting barrel cortex neurons, and that eventual deficits in inhibition may have differential effects on the excitation/inhibition balance in adapting and non-adapting neurons.
Collapse
|
45
|
Zhu L, Wang L, Ju F, Ran Y, Wang C, Zhang S. Transient global cerebral ischemia induces rapid and sustained reorganization of synaptic structures. J Cereb Blood Flow Metab 2017; 37:2756-2767. [PMID: 27798269 PMCID: PMC5536786 DOI: 10.1177/0271678x16674736] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Ischemia can cause rapid neuronal damage. Previous studies have suggested that synaptic structures and cortical functions can be rescued if therapeutic interventions are applied in time, but the structural basis for this resilience remains incompletely understood. Here, we investigated the restoration of synaptic structures and postischemic plasticity of dendritic spines in the somatosensory cortices of mice by taking advantage of a reversible global cerebral ischemia model. Intravital two-photon imaging revealed that although dendritic structures were rapidly distorted after global ischemia, only a small percentage of spines were actually lost after transient ischemia. Electron microscopy indicated that most presynaptic electron-dense structures were still apposed to postsynaptic densities, and that the majority of disrupted synaptic structures were rapidly reinstated following reperfusion after transient ischemia. Repeated imaging suggested that restored dendrites survived the initial ischemia -reperfusion challenge. Importantly, spines on the restored dendrites underwent a rapid and sustained structural reorganization following transient ischemia. These findings suggested that disrupted synapses during transient ischemia could be rapidly restored after ischemia/reperfusion, and that restored dendritic structures remained plastic to rebuild the cortical network.
Collapse
Affiliation(s)
- Lirui Zhu
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Lei Wang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Furong Ju
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Yanli Ran
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Cong Wang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Shengxiang Zhang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|
46
|
Tennant KA, Taylor SL, White ER, Brown CE. Optogenetic rewiring of thalamocortical circuits to restore function in the stroke injured brain. Nat Commun 2017. [PMID: 28643802 PMCID: PMC5490053 DOI: 10.1038/ncomms15879] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
To regain sensorimotor functions after stroke, surviving neural circuits must reorganize and form new connections. Although the thalamus is critical for processing and relaying sensory information to the cortex, little is known about how stroke affects the structure and function of these connections, or whether a therapeutic approach targeting these circuits can improve recovery. Here we reveal with in vivo calcium imaging that stroke in somatosensory cortex dampens the excitability of surviving thalamocortical circuits. Given this deficit, we hypothesized that chronic transcranial window optogenetic stimulation of thalamocortical axons could facilitate recovery. Using two-photon imaging, we show that optogenetic stimulation promotes the formation of new and stable thalamocortical synaptic boutons, without impacting axon branch dynamics. Stimulation also enhances the recovery of somatosensory cortical circuit function and forepaw sensorimotor abilities. These results demonstrate that an optogenetic approach can rewire thalamocortical circuits and restore function in the damaged brain. Stroke recovery requires circuit reorganization and therapeutic efforts have focused on rewiring cortical circuits after stroke, but what about thalamic inputs? Here, the authors examine how thalamocortical axons are affected by stroke and use optogenetic stimulation to promote recovery.
Collapse
Affiliation(s)
- Kelly A Tennant
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada V8P 5C2
| | - Stephanie L Taylor
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada V8P 5C2
| | - Emily R White
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada V8P 5C2
| | - Craig E Brown
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada V8P 5C2.,Department of Biology, University of Victoria, Victoria, British Columbia, Canada V8P 5C2.,Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
47
|
Electroacupuncture Regulates Hippocampal Synaptic Plasticity via miR-134-Mediated LIMK1 Function in Rats with Ischemic Stroke. Neural Plast 2017; 2017:9545646. [PMID: 28116173 PMCID: PMC5237739 DOI: 10.1155/2017/9545646] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 11/03/2016] [Indexed: 11/17/2022] Open
Abstract
MircoRNAs (miRs) have been implicated in learning and memory, by regulating LIM domain kinase (LIMK1) to induce synaptic-dendritic plasticity. The study aimed to investigate whether miRNAs/LIMK1 signaling was involved in electroacupuncture- (EA-) mediated synaptic-dendritic plasticity in a rat model of middle cerebral artery occlusion induced cognitive deficit (MICD). Compared to untreatment or non-acupoint-EA treatment, EA at DU20 and DU24 acupoints could shorten escape latency and increase the frequency of crossing platform in Morris water maze test. T2-weighted imaging showed that the MICD rat brain lesions were located in cortex, hippocampus, corpus striatum, and thalamus regions and injured volumes were reduced after EA. Furthermore, we found that the density of dendritic spine and the number of synapses in the hippocampal CA1 pyramidal cells were obviously reduced at Day 14 after MICD. However, synaptic-dendritic loss could be rescued after EA. Moreover, the synaptic-dendritic plasticity was associated with increases of the total LIMK1 and phospho-LIMK1 levels in hippocampal CA1 region, wherein EA decreased the expression of miR-134, negatively regulating LIMK1 to enhance synaptic-dendritic plasticity. Therefore, miR-134-mediated LIMK1 was involved in EA-induced hippocampal synaptic plasticity, which served as a contributor to improving learning and memory during the recovery stage of ischemic stroke.
Collapse
|
48
|
Blood-brain barrier breakdown and neovascularization processes after stroke and traumatic brain injury. Curr Opin Neurol 2016; 28:556-64. [PMID: 26402408 DOI: 10.1097/wco.0000000000000248] [Citation(s) in RCA: 220] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Angiogenesis or vascular reorganization plays a role in recovery after stroke and traumatic brain injury (TBI). In this review, we have focused on two major events that occur during stroke and TBI from a vascular perspective - what is the process and time course of blood-brain barrier (BBB) breakdown? and how does the surrounding vasculature recover and facilitate repair? RECENT FINDINGS Despite differences in the primary injury, the BBB changes overlap between stroke and TBI. Disruption of BBB involves a series of events: formation of caveolae, trans and paracellular disruption, tight junction breakdown and vascular disruption. Confounding factors that need careful assessment and standardization are the severity, duration and extent of the stroke and TBI that influences BBB disruption. Vascular repair proceeds through long-term neovascularization processes: angiogenesis, arteriogenesis and vasculogenesis. Enhancing each of these processes may impart beneficial effects in endogenous recovery. SUMMARY Our understanding of BBB breakdown acutely after the cerebrovascular injury has come a long way; however, we lack a clear understanding of the course of BBB disruption and BBB recovery and the evolution of individual cellular events associated with BBB change. Neovascularization responses have been widely studied in stroke for their role in functional recovery but the role of vascular reorganization after TBI in recovery is much less defined.
Collapse
|
49
|
He W, Au-Yeung SYS, Mak M, Leung TWH, Leung H, Wong LKS. The potential synergism by combining external counterpulsation with intermittent theta burst stimulation in post-stroke motor function recovery. Med Hypotheses 2016; 93:140-2. [PMID: 27372874 DOI: 10.1016/j.mehy.2016.05.024] [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: 02/27/2016] [Accepted: 05/22/2016] [Indexed: 11/16/2022]
Abstract
Upper limb weakness and incoordination is a common disability following ischemic stroke. Previous studies have showed that the single application of external counterpulsation (ECP) and intermittent theta burst stimulation (iTBS) can effectively enhance the cortical motor excitability and facilitate recovery. However, it remains uncertain if sequential application of these therapies would further augment the recovery. We hypothesize a synergistic effect of ECP followed by iTBS to upper limb function may happen through improvements in both cerebral perfusion and neuron excitability.
Collapse
Affiliation(s)
- Weijia He
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | | | - Margaret Mak
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong
| | - Thomas Wai Hong Leung
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong; Division of Neurology, Prince of Wales Hospital, Shatin, Hong Kong
| | - Howan Leung
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong; Division of Neurology, Prince of Wales Hospital, Shatin, Hong Kong
| | - Lawrence Ka Sing Wong
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong; Division of Neurology, Prince of Wales Hospital, Shatin, Hong Kong.
| |
Collapse
|
50
|
Reeson P, Jeffery A, Brown CE. Illuminating the Effects of Stroke on the Diabetic Brain: Insights From Imaging Neural and Vascular Networks in Experimental Animal Models. Diabetes 2016; 65:1779-88. [PMID: 27329953 DOI: 10.2337/db16-0064] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 03/31/2016] [Indexed: 11/13/2022]
Abstract
Type 1 diabetes is known to cause circulatory problems in the eyes, heart, and limbs, and the brain is no exception. Because of the insidious effects of diabetes on brain circulation, patients with diabetes are two to four times more likely to have an ischemic stroke and are less likely to regain functions that are lost. To provide a more mechanistic understanding of this clinically significant problem, imaging studies have focused on how stroke affects neural and vascular networks in experimental models of type 1 diabetes. The emerging picture is that diabetes leads to maladaptive changes in the cerebrovascular system that ultimately limit neuronal rewiring and recovery of functions after stroke. At the cellular and systems level, diabetes is associated with abnormal cerebral blood flow in surviving brain regions and greater disruption of the blood-brain barrier. The abnormal vascular responses to stroke can be partly attributed to aberrant vascular endothelial growth factor (VEGF) signaling because genetic or pharmacological inhibition of VEGF signaling can mitigate vascular dysfunction and improve stroke recovery in diabetic animals. These experimental studies offer new insights and strategies for optimizing stroke recovery in diabetic populations.
Collapse
Affiliation(s)
- Patrick Reeson
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Andrew Jeffery
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Craig E Brown
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada Department of Biology, University of Victoria, Victoria, British Columbia, Canada Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|