1
|
Li W, George P, Azadian MM, Ning M, Dhand A, Cramer SC, Carmichael ST, Lo EH. Changing genes, cells and networks to reprogram the brain after stroke. Nat Neurosci 2025; 28:1130-1145. [PMID: 40456908 DOI: 10.1038/s41593-025-01981-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 03/04/2025] [Indexed: 06/11/2025]
Abstract
Important advances have been made in reperfusion therapies for acute ischemic stroke. However, a majority of patients are either ineligible for or do not respond to treatments and continue to have considerable functional deficits. Stroke results in a pathological disruption of the neurovascular unit (NVU) that involves blood-brain barrier leakage, glial activation, neuronal damage and chronic inflammation, all of which create a microenvironment that hinders recovery. Therefore, finding ways to promote central nervous system recovery remains the holy grail of stroke research. Here we propose a conceptual framework to synthesize recent progress in the field, which is currently dispersed and disconnected in the literature. We suggest that stroke recovery requires an integrated reprogramming process throughout the brain that occurs at multiple levels, including changes in gene expression, endogenous cellular transdifferentiation within the NVU, and reorganization of larger-scale neural and social networks.
Collapse
Affiliation(s)
- Wenlu Li
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Zhejiang Key Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China.
| | - Paul George
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Matine M Azadian
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - MingMing Ning
- Clinical Proteomics Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Amar Dhand
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven C Cramer
- Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Lemarchand E, Grayston A, Wong R, Rogers M, Ouvrier B, Llewellyn B, Webb F, Lénárt N, Dénes Á, Brough D, Allan SM, Bix GJ, Pinteaux E. Selective deletion of interleukin-1 alpha in microglia does not modify acute outcome but may regulate neurorepair processes after experimental ischemic stroke. J Cereb Blood Flow Metab 2025:271678X251323371. [PMID: 40110693 PMCID: PMC11926816 DOI: 10.1177/0271678x251323371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/22/2025] [Accepted: 02/09/2025] [Indexed: 03/22/2025]
Abstract
Inflammation is a key contributor to stroke pathogenesis and exacerbates brain damage leading to poor outcome. Interleukin-1 (IL-1) is an important regulator of post-stroke inflammation, and blocking its actions is beneficial in pre-clinical stroke models and safe in the clinical setting. However, the distinct roles of the two major IL-1 receptor type 1 agonists, IL-1α and IL-1β, and the specific role of IL-1α in ischemic stroke remain largely unknown. Here we show that IL-1α and IL-1β have different spatio-temporal expression profiles in the brain after experimental stroke, with early microglial IL-1α expression (4 h) and delayed IL-1β expression in infiltrated neutrophils and a small microglial subset (24-72 h). We examined for the first time the specific role of microglial-derived IL-1α in experimental permanent and transient ischemic stroke through microglial-specific tamoxifen-inducible Cre-loxP-mediated recombination. Microglial IL-1α deletion did not influence acute outcome after ischemic stroke. However, microglial IL-1α knock out (KO) mice showed reduced peri-infarct vessel density and reactive astrogliosis at 14 days post-stroke, alongside long-term impaired functional recovery. Our study identifies for the first time a critical role for microglial IL-1α on post-stroke neurorepair and recovery, highlighting the importance of targeting specific IL-1 mechanisms in brain injury to develop effective therapies.
Collapse
Affiliation(s)
- Eloïse Lemarchand
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| | - Alba Grayston
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| | - Raymond Wong
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| | - Miyako Rogers
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| | - Blake Ouvrier
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, Orleans, LA, USA
| | - Benjamin Llewellyn
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| | - Freddie Webb
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| | - Nikolett Lénárt
- “Momentum” Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Ádám Dénes
- “Momentum” Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - David Brough
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| | - Stuart M Allan
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| | - Gregory J Bix
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, Orleans, LA, USA
| | - Emmanuel Pinteaux
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health (FBMH), The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Northern Care Alliance NHS Foundation Trust, The Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
3
|
Jones TA, Nemchek V, Fracassi M. Experience-driven competition in neural reorganization after stroke. J Physiol 2025; 603:737-757. [PMID: 39476290 PMCID: PMC11785499 DOI: 10.1113/jp285565] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/27/2024] [Indexed: 02/01/2025] Open
Abstract
Behavioural experiences interact with regenerative responses to shape patterns of neural reorganization after stroke. This review is focused on the competitive nature of these behavioural experience effects. Interactions between learning-related plasticity and regenerative reactions have been found to underlie the establishment of new compensatory behaviours and the efficacy of motor rehabilitative training in rodent stroke models. Learning in intact brains depends on competitive and cooperative mechanisms of synaptic plasticity. Synapses are added in response to learning and selectively maintained and strengthened via activity-dependent competition. Long-term memories for experiences that occur closely in time can be weakened or enhanced by competitive or cooperative interactions in the time-dependent process of stabilizing synaptic changes. Rodent stroke model findings suggest that compensatory reliance on the non-paretic hand after stroke can shape and stabilize synaptic reorganization patterns in both hemispheres, to compete with the capacity for experiences of the paretic side to do so. However, the competitive edge of the non-paretic side can be countered by overlapping experiences of the paretic hand, and might even be shifted in a cooperative direction with skilfully coordinated bimanual experience. Advances in the basic understanding of learning-related synaptic competition are helping to inform the basis of experience-dependent variations in stroke outcome.
Collapse
Affiliation(s)
- Theresa A. Jones
- Psychology Department & Institute for Neuroscience, University of Texas at Austin
| | - Victoria Nemchek
- Psychology Department & Institute for Neuroscience, University of Texas at Austin
| | - Michela Fracassi
- Psychology Department & Institute for Neuroscience, University of Texas at Austin
| |
Collapse
|
4
|
Hagemann N, Qi Y, Mohamud Yusuf A, Li A, Zhang X, Spangenberg P, Squire A, Doeppner TR, Jin F, Zhao S, Chen J, Mosig A, Gunzer M, Hermann DM. Arterial specification precedes microvascular restitution in the peri-infarct cortex that is driven by small microvessels. J Cereb Blood Flow Metab 2025; 45:171-186. [PMID: 39113408 PMCID: PMC11571960 DOI: 10.1177/0271678x241270407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/10/2024] [Accepted: 06/21/2024] [Indexed: 11/20/2024]
Abstract
Evaluation of microvascular networks was impeded until recently by the need of histological tissue sectioning, which precluded 3D analyses. Using light-sheet microscopy, we investigated microvascular network characteristics in the peri-infarct cortex of mice 3-56 days after transient middle cerebral artery occlusion. In animal subgroups, the sphingosine-1-phosphate analog FTY720 (Fingolimod) was administered starting 24 hours post-ischemia. Light-sheet microscopy revealed a striking pattern of microvascular changes in the peri-infarct cortex, that is, a loss of microvessels, which was most prominent after 7 days and followed by the reappearance of microvessels over 56 days which revealed an increased branching point density and shortened branches. Using a novel AI-based image analysis algorithm we found that the length density of microvessels expressing the arterial specification marker α-smooth muscle actin markedly increased in the peri-infarct cortex already at 7 days post-ischemia. The length and branch density of small microvessels, but not of intermediate or large microvessels increased above pre-ischemic levels within 14-56 days. FTY720 increased the length and branch density of small microvessels. This study demonstrates long-term alterations of microvascular architecture post-ischemia indicative of increased collateralization most notably of small microvessels. Light-sheet microscopy will greatly advance the assessment of microvascular responses to restorative stroke therapies.
Collapse
Affiliation(s)
- Nina Hagemann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
| | - Yachao Qi
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
| | - Ayan Mohamud Yusuf
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
| | - AnRan Li
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
| | - Xiaoni Zhang
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
| | - Philippa Spangenberg
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Imaging Center Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Anthony Squire
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Imaging Center Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Thorsten R Doeppner
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
| | - Fengyan Jin
- Department of Hematology, First Hospital of Jilin University, Changchun, Jilin, China
| | - Shuo Zhao
- Leibniz-Institut für Analytische Wissenschaften (ISAS), Dortmund, Germany
| | - Jianxu Chen
- Leibniz-Institut für Analytische Wissenschaften (ISAS), Dortmund, Germany
| | - Axel Mosig
- Bioinformatics Group, Center for Protein Diagnostics, Ruhr-University, Bochum, Germany
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Imaging Center Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Leibniz-Institut für Analytische Wissenschaften (ISAS), Dortmund, Germany
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
| |
Collapse
|
5
|
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
|
6
|
Rust R, Nih LR, Liberale L, Yin H, El Amki M, Ong LK, Zlokovic BV. Brain repair mechanisms after cell therapy for stroke. Brain 2024; 147:3286-3305. [PMID: 38916992 PMCID: PMC11449145 DOI: 10.1093/brain/awae204] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/04/2024] [Accepted: 06/08/2024] [Indexed: 06/27/2024] Open
Abstract
Cell-based therapies hold great promise for brain repair after stroke. While accumulating evidence confirms the preclinical and clinical benefits of cell therapies, the underlying mechanisms by which they promote brain repair remain unclear. Here, we briefly review endogenous mechanisms of brain repair after ischaemic stroke and then focus on how different stem and progenitor cell sources can promote brain repair. Specifically, we examine how transplanted cell grafts contribute to improved functional recovery either through direct cell replacement or by stimulating endogenous repair pathways. Additionally, we discuss recently implemented preclinical refinement methods, such as preconditioning, microcarriers, genetic safety switches and universal (immune evasive) cell transplants, as well as the therapeutic potential of these pharmacologic and genetic manipulations to further enhance the efficacy and safety of cell therapies. By gaining a deeper understanding of post-ischaemic repair mechanisms, prospective clinical trials may be further refined to advance post-stroke cell therapy to the clinic.
Collapse
Affiliation(s)
- Ruslan Rust
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
| | - Lina R Nih
- Department of Brain Health, University of Nevada, Las Vegas, NV 89154, USA
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Hao Yin
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Mohamad El Amki
- Department of Neurology, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Lin Kooi Ong
- School of Health and Medical Sciences & Centre for Health Research, University of Southern Queensland, Toowoomba, QLD 4350, Australia
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
7
|
Seblani M, Brezun JM, Féron F, Hoquet T. Rethinking plasticity: Analysing the concept of "destructive plasticity" in the light of neuroscience definitions. Eur J Neurosci 2024; 60:4798-4812. [PMID: 39092545 DOI: 10.1111/ejn.16487] [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: 01/16/2024] [Revised: 06/19/2024] [Accepted: 07/17/2024] [Indexed: 08/04/2024]
Abstract
As a multilevel and multidisciplinary field, neuroscience is designed to interact with various branches of natural and applied sciences as well as with humanities and philosophy. The continental tradition in philosophy, particularly over the past 20 years, tended to establish strong connections with biology and neuroscience findings. This cross fertilization can however be impeded by conceptual intricacies, such as those surrounding the concept of plasticity. The use of this concept has broadened as scientists applied it to explore an ever-growing range of biological phenomena. Here, we examine the consequences of this ambiguity in an interdisciplinary context through the analysis of the concept of "destructive plasticity" in the philosophical writings of Catherine Malabou. The term "destructive plasticity" was coined by Malabou in 2009 to refer to all processes leading to psycho-cognitive and emotional alterations following traumatic or nontraumatic brain injuries or resulting from neurodevelopmental disorders. By comparing it with the neuroscientific definitions of plasticity, we discuss the epistemological obstacles and possibilities related to the integration of this concept into neuroscience. Improving interdisciplinary exchanges requires an advanced and sophisticated manipulation of neurobiological concepts. These concepts are not only intended to guide research programmes within neuroscience but also to organize and frame the dialogue between different theoretical backgrounds.
Collapse
Affiliation(s)
- Mostafa Seblani
- Institut des Sciences du Mouvement: Etienne-Jules MAREY (ISM), CNRS, Aix Marseille Univ, UMR 7287, Campus Scientifique de Luminy, Marseille Cedex 09, France
- Institute of NeuroPhysiopathology (INP), CNRS, Aix Marseille University, UMR 7051, Marseille Cedex 5, France
- Department of Philosophy, University Paris Nanterre, Nanterre Cedex, France
| | - Jean-Michel Brezun
- Institut des Sciences du Mouvement: Etienne-Jules MAREY (ISM), CNRS, Aix Marseille Univ, UMR 7287, Campus Scientifique de Luminy, Marseille Cedex 09, France
| | - François Féron
- Institute of NeuroPhysiopathology (INP), CNRS, Aix Marseille University, UMR 7051, Marseille Cedex 5, France
| | - Thierry Hoquet
- Department of Philosophy, University Paris Nanterre, Nanterre Cedex, France
| |
Collapse
|
8
|
Mihelic SA, Engelmann SA, Sadr M, Jafari CZ, Zhou A, Woods AL, Williamson MR, Jones TA, Dunn AK. Microvascular plasticity in mouse stroke model recovery: Anatomy statistics, dynamics measured by longitudinal in vivo two-photon angiography, network vectorization. J Cereb Blood Flow Metab 2024:271678X241270465. [PMID: 39113424 PMCID: PMC11572002 DOI: 10.1177/0271678x241270465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/19/2024] [Accepted: 06/23/2024] [Indexed: 11/20/2024]
Abstract
This manuscript quantitatively investigates remodeling dynamics of the cortical microvascular network (thousands of connected capillaries) following photothrombotic ischemia (cubic millimeter volume, imaged weekly) using a novel in vivo two-photon angiography and high throughput vascular vectorization method. The results suggest distinct temporal patterns of cerebrovascular plasticity, with acute remodeling peaking at one week post-stroke. The network architecture then gradually stabilizes, returning to a new steady state after four weeks. These findings align with previous literature on neuronal plasticity, highlighting the correlation between neuronal and neurovascular remodeling. Quantitative analysis of neurovascular networks using length- and strand-based statistical measures reveals intricate changes in network anatomy and topology. The distance and strand-length statistics show significant alterations, with a peak of plasticity observed at one week post-stroke, followed by a gradual return to baseline. The orientation statistic plasticity peaks at two weeks, gradually approaching the (conserved across subjects) stroke signature. The underlying mechanism of the vascular response (angiogenesis vs. tissue deformation), however, is yet unexplored. Overall, the combination of chronic two-photon angiography, vascular vectorization, reconstruction/visualization, and statistical analysis enables both qualitative and quantitative assessments of neurovascular remodeling dynamics, demonstrating a method for investigating cortical microvascular network disorders and the therapeutic modes of action thereof.
Collapse
Affiliation(s)
- Samuel A Mihelic
- Biomedical Engineering Department, University of Texas at Austin, Austin, TX, USA
| | - Shaun A Engelmann
- Biomedical Engineering Department, University of Texas at Austin, Austin, TX, USA
| | - Mahdi Sadr
- Biomedical Engineering Department, University of Texas at Austin, Austin, TX, USA
| | - Chakameh Z Jafari
- Biomedical Engineering Department, University of Texas at Austin, Austin, TX, USA
| | - Annie Zhou
- Biomedical Engineering Department, University of Texas at Austin, Austin, TX, USA
| | - Aaron L Woods
- Biomedical Engineering Department, University of Texas at Austin, Austin, TX, USA
| | | | - Theresa A Jones
- Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
| | - Andrew K Dunn
- Biomedical Engineering Department, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
9
|
Lemarchand E, Grayston A, Wong R, Rogers M, Ouvrier B, Llewellyn B, Webb F, Lénárt N, Denes A, Brough D, Allan SM, Bix GJ, Pinteaux E. Selective deletion of interleukin-1 alpha in microglia does not modify acute outcome but regulates neurorepair processes after experimental ischemic stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.16.580635. [PMID: 38585834 PMCID: PMC10996562 DOI: 10.1101/2024.02.16.580635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Inflammation is a key contributor to stroke pathogenesis and exacerbates brain damage leading to poor outcome. Interleukin-1 (IL-1) is an important regulator of post-stroke inflammation, and blocking its actions is beneficial in pre-clinical stroke models and safe in the clinical setting. However, the distinct roles of the two major IL-1 receptor type 1 agonists, IL-1α and IL-1β, and the specific role of IL-1α in ischemic stroke remain largely unknown. Here we show that IL-1α and IL-1β have different spatio-temporal expression profiles in the brain after experimental stroke, with early microglial IL-1α expression (4 h) and delayed IL-1β expression in infiltrated neutrophils and a small microglial subset (24-72 h). We examined for the first time the specific role of microglial-derived IL-1α in experimental permanent and transient ischemic stroke through microglial-specific tamoxifen-inducible Cre-loxP-mediated recombination. Microglial IL-1α deletion did not influence acute brain damage, cerebral blood flow, IL-1β expression, neutrophil infiltration, microglial nor endothelial activation after ischemic stroke. However, microglial IL-1α knock out (KO) mice showed reduced peri-infarct vessel density and reactive astrogliosis at 14 days post-stroke, alongside long-term impaired functional recovery. Our study identifies for the first time a critical role for microglial IL-1α on neurorepair and functional recovery after stroke, highlighting the importance of targeting specific IL-1 mechanisms in brain injury to develop more effective therapies.
Collapse
|
10
|
Michór P, Renardson L, Li S, Boltze J. Neurorestorative Approaches for Ischemic StrokeChallenges, Opportunities, and Recent Advances. Neuroscience 2024; 550:69-78. [PMID: 38763225 DOI: 10.1016/j.neuroscience.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/21/2024]
Abstract
Despite recent advances in acute stroke management, most patients experiencing a stroke will suffer from residual brain damage and functional impairment. Addressing those residual deficits would require neurorestoration, i.e., rebuilding brain tissue to repair the structural brain damage caused by stroke. However, there are major pathobiological, anatomical and technological hurdles making neurorestorative approaches remarkably challenging, and true neurorestoration after larger ischemic lesions could not yet be achieved. On the other hand, there has been steady advancement in our understanding of the limits of tissue regeneration in the adult mammalian brain as well as of the fundamental organization of brain tissue growth during embryo- and ontogenesis. This has been paralleled by the development of novel animal models to study stroke, advancement of biomaterials that can be used to support neurorestoration, and in stem cell technologies. This review gives a detailed explanation of the major hurdles so far preventing the achievement of neurorestoration after stroke. It will also describe novel concepts and advancements in biomaterial science, brain organoid culturing, and animal modeling that may enable the investigation of post-stroke neurorestorative approaches in translationally relevant setups. Finally, there will be a review of recent achievements in experimental studies that have the potential to be the starting point of research and development activities that may eventually bring post-stroke neurorestoration within reach.
Collapse
Affiliation(s)
- Paulina Michór
- University of Warwick, School of Life Sciences, Coventry CV4 7AL, United Kingdom
| | - Lydia Renardson
- University of Warwick, Warwick Medical School, Coventry CV4 7AL, United Kingdom
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Johannes Boltze
- University of Warwick, School of Life Sciences, Coventry CV4 7AL, United Kingdom.
| |
Collapse
|
11
|
Zhang C, Jamshidi M, Delafontaine-Martel P, Linninger AA, Lesage F. Evaluation of cerebral microcirculation in a mouse model of systemic inflammation. NEUROPHOTONICS 2024; 11:035003. [PMID: 39011517 PMCID: PMC11249390 DOI: 10.1117/1.nph.11.3.035003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 07/17/2024]
Abstract
Significance Perturbations in the microcirculatory system have been observed in neurological conditions, such as Alzheimer's disease or systemic inflammation. However, changes occurring at the level of the capillary are difficult to translate to biomarkers that could be measured macroscopically. Aim We aim to evaluate whether transit time changes reflect capillary stalling and to what degree. Approach We employ a combined spectral optical coherence tomography (OCT) and fluorescence optical imaging (FOI) system to investigate the relation between capillary stalling and transit time in a mouse model of systemic inflammation induced by intraperitoneal injection of lipopolysaccharide. Angiograms are obtained using OCT, and fluorescence signal images are acquired by the FOI system upon intravenous injection of fluorescein isothiocyanate via a catheter inserted into the tail vein. Results Our findings reveal that lipopolysaccharide (LPS) administration significantly increases both the percentage and duration of capillary stalling compared to mice receiving a 0.9% saline injection. Moreover, LPS-induced mice exhibit significantly prolonged arteriovenous transit time compared to control mice. Conclusions These observations suggest that capillary stalling, induced by inflammation, modulates cerebral mean transit time, a measure that has translational potential.
Collapse
Affiliation(s)
- Cong Zhang
- Polytechnique Montreal, Department of Electrical Engineering, Montreal, Quebec, Canada
- Montreal Heart Institute, Research center, Montreal, Quebec, Canada
| | - Mohammad Jamshidi
- University of Illinois at Chicago, Department of Biomedical Engineering, Chicago, Illinois, United States
| | - Patrick Delafontaine-Martel
- Polytechnique Montreal, Department of Electrical Engineering, Montreal, Quebec, Canada
- Montreal Heart Institute, Research center, Montreal, Quebec, Canada
| | - Andreas A Linninger
- University of Illinois at Chicago, Department of Biomedical Engineering, Chicago, Illinois, United States
- University of Illinois at Chicago, Department of Neurosurgery, Chicago, Illinois, United States
| | - Frédéric Lesage
- Polytechnique Montreal, Department of Electrical Engineering, Montreal, Quebec, Canada
- Montreal Heart Institute, Research center, Montreal, Quebec, Canada
| |
Collapse
|
12
|
Lam DV, Javadekar A, Patil N, Yu M, Li L, Menendez DM, Gupta AS, Capadona JR, Shoffstall AJ. Corrigendum to "Platelets and Hemostatic Proteins are Co-Localized with Chronic Neuroinflammation Surrounding Implanted Intracortical Microelectrodes" [Acta Biomaterialia. Volume 166, August 2023, Pages 278-290]. Acta Biomater 2024; 182:303-308. [PMID: 38845260 PMCID: PMC11295673 DOI: 10.1016/j.actbio.2024.05.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/19/2024]
Affiliation(s)
- Danny V Lam
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Anisha Javadekar
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | | | - Marina Yu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Longshun Li
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Dhariyat M Menendez
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Andrew J Shoffstall
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA.
| |
Collapse
|
13
|
Li Y, Xue W, Li S, Cui L, Gao Y, Li L, Chen R, Zhang X, Xu R, Jiang W, Zhang X, Wang L. Salidroside promotes angiogenesis after cerebral ischemia in mice through Shh signaling pathway. Biomed Pharmacother 2024; 174:116625. [PMID: 38643543 DOI: 10.1016/j.biopha.2024.116625] [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: 12/19/2023] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 04/23/2024] Open
Abstract
AIMS The purpose of this study was to explore the impacts of salidroside on vascular regeneration, vascular structural changes and long-term neurological recuperation following cerebral ischemia and its possible mechanism. MAIN METHODS From Day 1 to Day 28, young male mice with middle cerebral artery blockage received daily doses of salidroside and measured neurological deficits. On the 7th day after stroke, the volume of cerebral infarction was determined using TTC and HE staining. Microvascular density, astrocyte coverage, angiogenesis and the expression of the Shh signaling pathway were detected by IF, qRTPCR and WB at 7, 14 and 28 days after stroke. Changes in blood flow, blood vessel density and diameter from stroke to 28 days were measured by the LSCI and TPMI. KEY FINDINGS Compared with the dMACO group, the salidroside treatment group significantly promoted the recovery of neurological function. Salidroside was found to enhance cerebral blood flow perfusion and reduce the infarct on the 7th day after stroke. From the 7th to the 28th day after stroke, salidroside treatment boosted the expression of CD31, CD31+/BrdU+, and GFAP in the cortex around the infarction site. On the 14th day after stroke, salidroside significantly enhanced the width and density of blood vessels. Salidroside increased the expression of histones and genes in the Shh signaling pathway during treatment, and this effect was weakened by the Shh inhibitor Cyclopamine. SIGNIFICANCE Salidroside can restore nerve function, improve cerebral blood flow, reduce cerebral infarction volume, increase microvessel density and promote angiogenesis via the Shh signaling pathway.
Collapse
Affiliation(s)
- Ying Li
- Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Weihong Xue
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Songyi Li
- Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Lili Cui
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Yuxiao Gao
- Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Linlin Li
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Rong Chen
- Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Xiao Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Renhao Xu
- Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Wei Jiang
- Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China.
| | - Lina Wang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China.
| |
Collapse
|
14
|
Protzmann J, Jung F, Jakobsson L, Fredriksson L. Analysis of ischemic stroke-mediated effects on blood-brain barrier properties along the arteriovenous axis assessed by intravital two-photon imaging. Fluids Barriers CNS 2024; 21:35. [PMID: 38622710 PMCID: PMC11017501 DOI: 10.1186/s12987-024-00537-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 04/03/2024] [Indexed: 04/17/2024] Open
Abstract
Early breach of the blood-brain barrier (BBB) and consequently extravasation of blood-borne substances into the brain parenchyma is a common hallmark of ischemic stroke. Although BBB breakdown is associated with an increased risk of cerebral hemorrhage and poor clinical prognosis, the cause and mechanism of this process are largely unknown. The aim of this study was to establish an imaging and analysis protocol which enables investigation of the dynamics of BBB breach in relation to hemodynamic properties along the arteriovenous axis. Using longitudinal intravital two-photon imaging following photothrombotic induction of ischemic stroke through a cranial window, we were able to study the response of the cerebral vasculature to ischemia, from the early critical hours to the days/weeks after the infarct. We demonstrate that disruption of the BBB and hemodynamic parameters, including perturbed blood flow, can be studied at single-vessel resolution in the three-dimensional space as early as 30 min after vessel occlusion. Further, we show that this protocol permits longitudinal studies on the response of individual blood vessels to ischemia over time, thus enabling detection of (maladaptive) vascular remodeling such as intussusception, angiogenic sprouting and entanglement of vessel networks. Taken together, this in vivo two-photon imaging and analysis protocol will be useful in future studies investigating the molecular and cellular mechanisms, and the spatial contribution, of BBB breach to disease progression which might ultimately aid the development of new and more precise treatment strategies for ischemic stroke.
Collapse
Affiliation(s)
- Jil Protzmann
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Solnavägen 9, Stockholm, Sweden, 17165
| | - Felix Jung
- Department of Neuroscience , Karolinska Institutet, Solnavägen 9, Stockholm, Sweden, 17165
| | - Lars Jakobsson
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Solnavägen 9, Stockholm, Sweden, 17165
| | - Linda Fredriksson
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Solnavägen 9, Stockholm, Sweden, 17165.
| |
Collapse
|
15
|
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
|
16
|
Hagemann N, Qi Y, Mohamud Yusuf A, Li A, Squire A, Tertel T, Giebel B, Ludewig P, Spangenberg P, Chen J, Mosig A, Gunzer M, Hermann DM. Microvascular Network Remodeling in the Ischemic Mouse Brain Defined by Light Sheet Microscopy. Arterioscler Thromb Vasc Biol 2024; 44:915-929. [PMID: 38357819 DOI: 10.1161/atvbaha.123.320339] [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: 10/27/2023] [Accepted: 02/01/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Until now, the analysis of microvascular networks in the reperfused ischemic brain has been limited due to tissue transparency challenges. METHODS Using light sheet microscopy, we assessed microvascular network remodeling in the striatum from 3 hours to 56 days post-ischemia in 2 mouse models of transient middle cerebral artery occlusion lasting 20 or 40 minutes, resulting in mild ischemic brain injury or brain infarction, respectively. We also examined the effect of a clinically applicable S1P (sphingosine-1-phosphate) analog, FTY720 (fingolimod), on microvascular network remodeling. RESULTS Over 56 days, we observed progressive microvascular degeneration in the reperfused striatum, that is, the lesion core, which was followed by robust angiogenesis after mild ischemic injury induced by 20-minute middle cerebral artery occlusion. However, more severe ischemic injury elicited by 40-minute middle cerebral artery occlusion resulted in incomplete microvascular remodeling. In both cases, microvascular networks did not return to their preischemic state but displayed a chronically altered pattern characterized by higher branching point density, shorter branches, higher unconnected branch density, and lower tortuosity, indicating enhanced network connectivity. FTY720 effectively increased microvascular length density, branching point density, and volume density in both models, indicating an angiogenic effect of this drug. CONCLUSIONS Utilizing light sheet microscopy together with automated image analysis, we characterized microvascular remodeling in the ischemic lesion core in unprecedented detail. This technology will significantly advance our understanding of microvascular restorative processes and pave the way for novel treatment developments in the stroke field.
Collapse
Affiliation(s)
- Nina Hagemann
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Germany (N.H., Y.Q., A.M.Y., A.L., D.M.H.)
| | - Yachao Qi
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Germany (N.H., Y.Q., A.M.Y., A.L., D.M.H.)
| | - Ayan Mohamud Yusuf
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Germany (N.H., Y.Q., A.M.Y., A.L., D.M.H.)
| | - AnRan Li
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Germany (N.H., Y.Q., A.M.Y., A.L., D.M.H.)
| | - Anthony Squire
- Institute for Experimental Immunology and Imaging and Imaging Center Essen, University Hospital Essen, University of Duisburg-Essen, Germany (A.S., P.S., M.G.)
| | - Tobias Tertel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Germany (T.T., B.G.)
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Germany (T.T., B.G.)
| | - Peter Ludewig
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Germany (P.L.)
| | - Philippa Spangenberg
- Institute for Experimental Immunology and Imaging and Imaging Center Essen, University Hospital Essen, University of Duisburg-Essen, Germany (A.S., P.S., M.G.)
| | - Jianxu Chen
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany (J.C., M.G.)
| | - Axel Mosig
- Bioinformatics Group, Faculty for Biology and Biotechnology and Center for Protein Diagnostics, Ruhr-University Bochum, Germany (A.M.)
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging and Imaging Center Essen, University Hospital Essen, University of Duisburg-Essen, Germany (A.S., P.S., M.G.)
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany (J.C., M.G.)
| | - Dirk M Hermann
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Germany (N.H., Y.Q., A.M.Y., A.L., D.M.H.)
| |
Collapse
|
17
|
Stavchansky VV, Yuzhakov VV, Sevan'kaeva LE, Fomina NK, Koretskaya AE, Denisova AE, Mozgovoy IV, Gubsky LV, Filippenkov IB, Myasoedov NF, Limborska SA, Dergunova LV. Melanocortin Derivatives Induced Vascularization and Neuroglial Proliferation in the Rat Brain under Conditions of Cerebral Ischemia. Curr Issues Mol Biol 2024; 46:2071-2092. [PMID: 38534749 DOI: 10.3390/cimb46030133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/28/2024] Open
Abstract
Stroke remains the second leading cause of death worldwide. The development of new therapeutic agents focused on restoring vascular function and neuroprotection of viable tissues is required. In this study the neuroprotective activity of melanocortin-like ACTH(4-7)PGP and ACTH(6-9)PGP peptides was investigated in rat brain at 24 h after transient middle cerebral artery occlusion (tMCAO). The severity of ischemic damage, changes in the proliferative activity of neuroglial cells and vascularization of rat brain tissue were analyzed. The administration of peptides resulted in a significant increase in the volume density of neurons in the perifocal zone of infarction compared to rats subjected to ischemia and receiving saline. Immunohistochemical analysis of the proliferative activity of neuroglia cells using PCNA antibodies showed a significant increase in the number of proliferating cells in the penumbra and in the intact cerebral cortex of rats receiving peptide treatment. The effect of peptides on vascularization was examined using CD31 antibodies under tMCAO conditions, revealing a significant increase in the volume density of vessels and their sizes in the penumbra after administration of ACTH(4-7)PGP and ACTH(6-9)PGP. These findings confirm the neuroprotective effect of peptides due to the activation of neuroglia proliferation and the enhancement of collateral blood flow.
Collapse
Affiliation(s)
- Vasily V Stavchansky
- National Research Center "Kurchatov Institute", Kurchatov Sq. 2, Moscow 123182, Russia
| | - Vadim V Yuzhakov
- A. Tsyb Medical Radiological Research Center-Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Koroleva Str. 4B, Obninsk 249036, Russia
| | - Larisa E Sevan'kaeva
- A. Tsyb Medical Radiological Research Center-Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Koroleva Str. 4B, Obninsk 249036, Russia
| | - Natalia K Fomina
- A. Tsyb Medical Radiological Research Center-Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Koroleva Str. 4B, Obninsk 249036, Russia
| | - Anastasia E Koretskaya
- A. Tsyb Medical Radiological Research Center-Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Koroleva Str. 4B, Obninsk 249036, Russia
| | - Alina E Denisova
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, Ostrovitianov Str. 1, Moscow 117997, Russia
| | - Ivan V Mozgovoy
- National Research Center "Kurchatov Institute", Kurchatov Sq. 2, Moscow 123182, Russia
| | - Leonid V Gubsky
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, Ostrovitianov Str. 1, Moscow 117997, Russia
| | - Ivan B Filippenkov
- National Research Center "Kurchatov Institute", Kurchatov Sq. 2, Moscow 123182, Russia
| | - Nikolay F Myasoedov
- National Research Center "Kurchatov Institute", Kurchatov Sq. 2, Moscow 123182, Russia
| | - Svetlana A Limborska
- National Research Center "Kurchatov Institute", Kurchatov Sq. 2, Moscow 123182, Russia
| | - Lyudmila V Dergunova
- National Research Center "Kurchatov Institute", Kurchatov Sq. 2, Moscow 123182, Russia
| |
Collapse
|
18
|
Goodman GW, Do TH, Tan C, Ritzel RM. Drivers of Chronic Pathology Following Ischemic Stroke: A Descriptive Review. Cell Mol Neurobiol 2023; 44:7. [PMID: 38112809 PMCID: PMC11391890 DOI: 10.1007/s10571-023-01437-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/25/2023] [Indexed: 12/21/2023]
Abstract
Stroke is the third leading cause of death and long-term disability in the world. Considered largely a disease of aging, its global economic and healthcare burden is expected to rise as more people survive into advanced age. With recent advances in acute stroke management, including the expansion of time windows for treatment with intravenous thrombolysis and mechanical thrombectomy, we are likely to see an increase in survival rates. It is therefore critically important to understand the complete pathophysiology of ischemic stroke, both in the acute and subacute stages and during the chronic phase in the months and years following an ischemic event. One of the most clinically relevant aspects of the chronic sequelae of stroke is its extended negative effect on cognition. Cognitive impairment may be related to the deterioration and dysfunctional reorganization of white matter seen at later timepoints after stroke, as well as ongoing progressive neurodegeneration. The vasculature of the brain also undergoes significant insult and remodeling following stroke, undergoing changes which may further contribute to chronic stroke pathology. While inflammation and the immune response are well established drivers of acute stroke pathology, the chronicity and functional role of innate and adaptive immune responses in the post-ischemic brain and in the peripheral environment remain largely uncharacterized. In this review, we summarize the current literature on post-stroke injury progression, its chronic pathological features, and the putative secondary injury mechanisms underlying the development of cognitive impairment and dementia. We present findings from clinical and experimental studies and discuss the long-term effects of ischemic stroke on both brain anatomy and functional outcome. Identifying mechanisms that occur months to years after injury could lead to treatment strategies in the chronic phase of stroke to help mitigate stroke-associated cognitive decline in patients.
Collapse
Affiliation(s)
- Grant W Goodman
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Trang H Do
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chunfeng Tan
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Rodney M Ritzel
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
19
|
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
|
20
|
Suo Q, Deng L, Chen T, Wu S, Qi L, Liu Z, He T, Tian HL, Li W, Tang Y, Yang GY, Zhang Z. Optogenetic Activation of Astrocytes Reduces Blood-Brain Barrier Disruption via IL-10 In Stroke. Aging Dis 2023; 14:1870-1886. [PMID: 37196130 PMCID: PMC10529757 DOI: 10.14336/ad.2023.0226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 02/26/2023] [Indexed: 05/19/2023] Open
Abstract
Optogenetics has been used to regulate astrocyte activity and modulate neuronal function after brain injury. Activated astrocytes regulate blood-brain barrier functions and are thereby involved in brain repair. However, the effect and molecular mechanism of optogenetic-activated astrocytes on the change in barrier function in ischemic stroke remain obscure. In this study, adult male GFAP-ChR2-EYFP transgenic Sprague-Dawley rats were stimulated by optogenetics at 24, 36, 48, and 60 h after photothrombotic stroke to activate ipsilateral cortical astrocytes. The effects of activated astrocytes on barrier integrity and the underlying mechanisms were explored using immunostaining, western blotting, RT-qPCR, and shRNA interference. Neurobehavioral tests were performed to evaluate therapeutic efficacy. The results demonstrated that IgG leakage, gap formation of tight junction proteins, and matrix metallopeptidase 2 expression were reduced after optogenetic activation of astrocytes (p<0.05). Moreover, photo-stimulation of astrocytes protected neurons against apoptosis and improved neurobehavioral outcomes in stroke rats compared to controls (p<0.05). Notably, interleukin-10 expression in optogenetic-activated astrocytes significantly increased after ischemic stroke in rats. Inhibition of interleukin-10 in astrocytes compromised the protective effects of optogenetic-activated astrocytes (p<0.05). We found for the first time that interleukin-10 derived from optogenetic-activated astrocytes protected blood-brain barrier integrity by decreasing the activity of matrix metallopeptidase 2 and attenuated neuronal apoptosis, which provided a novel therapeutic approach and target in the acute stage of ischemic stroke.
Collapse
Affiliation(s)
- Qian Suo
- Shanghai Jiao Tong Affiliated Sixth People’s Hospital, and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Lidong Deng
- Shanghai Jiao Tong Affiliated Sixth People’s Hospital, and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Tingting Chen
- Shanghai Jiao Tong Affiliated Sixth People’s Hospital, and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Shengju Wu
- Shanghai Jiao Tong Affiliated Sixth People’s Hospital, and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Lin Qi
- Shanghai Jiao Tong Affiliated Sixth People’s Hospital, and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Ze Liu
- Shanghai Jiao Tong Affiliated Sixth People’s Hospital, and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Tingting He
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Heng-Li Tian
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Wanlu Li
- Shanghai Jiao Tong Affiliated Sixth People’s Hospital, and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Yaohui Tang
- Shanghai Jiao Tong Affiliated Sixth People’s Hospital, and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Guo-Yuan Yang
- Shanghai Jiao Tong Affiliated Sixth People’s Hospital, and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Zhijun Zhang
- Shanghai Jiao Tong Affiliated Sixth People’s Hospital, and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
21
|
Wu X, Li JR, Fu Y, Chen DY, Nie H, Tang ZP. From static to dynamic: live observation of the support system after ischemic stroke by two photon-excited fluorescence laser-scanning microscopy. Neural Regen Res 2023; 18:2093-2107. [PMID: 37056116 PMCID: PMC10328295 DOI: 10.4103/1673-5374.369099] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/21/2022] [Accepted: 01/13/2023] [Indexed: 02/17/2023] Open
Abstract
Ischemic stroke is one of the most common causes of mortality and disability worldwide. However, treatment efficacy and the progress of research remain unsatisfactory. As the critical support system and essential components in neurovascular units, glial cells and blood vessels (including the blood-brain barrier) together maintain an optimal microenvironment for neuronal function. They provide nutrients, regulate neuronal excitability, and prevent harmful substances from entering brain tissue. The highly dynamic networks of this support system play an essential role in ischemic stroke through processes including brain homeostasis, supporting neuronal function, and reacting to injuries. However, most studies have focused on postmortem animals, which inevitably lack critical information about the dynamic changes that occur after ischemic stroke. Therefore, a high-precision technique for research in living animals is urgently needed. Two-photon fluorescence laser-scanning microscopy is a powerful imaging technique that can facilitate live imaging at high spatiotemporal resolutions. Two-photon fluorescence laser-scanning microscopy can provide images of the whole-cortex vascular 3D structure, information on multicellular component interactions, and provide images of structure and function in the cranial window. This technique shifts the existing research paradigm from static to dynamic, from flat to stereoscopic, and from single-cell function to multicellular intercommunication, thus providing direct and reliable evidence to identify the pathophysiological mechanisms following ischemic stroke in an intact brain. In this review, we discuss exciting findings from research on the support system after ischemic stroke using two-photon fluorescence laser-scanning microscopy, highlighting the importance of dynamic observations of cellular behavior and interactions in the networks of the brain's support systems. We show the excellent application prospects and advantages of two-photon fluorescence laser-scanning microscopy and predict future research developments and directions in the study of ischemic stroke.
Collapse
Affiliation(s)
- Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jia-Rui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yu Fu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Dan-Yang Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hao Nie
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhou-Ping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
22
|
Lam DV, Javadekar A, Patil N, Yu M, Li L, Menendez DM, Gupta AS, Capadona JR, Shoffstall AJ. Platelets and hemostatic proteins are co-localized with chronic neuroinflammation surrounding implanted intracortical microelectrodes. Acta Biomater 2023; 166:278-290. [PMID: 37211307 PMCID: PMC10330779 DOI: 10.1016/j.actbio.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/13/2023] [Accepted: 05/02/2023] [Indexed: 05/23/2023]
Abstract
Intracortical microelectrodes induce vascular injury upon insertion into the cortex. As blood vessels rupture, blood proteins and blood-derived cells (including platelets) are introduced into the 'immune privileged' brain tissues at higher-than-normal levels, passing through the damaged blood-brain barrier. Blood proteins adhere to implant surfaces, increasing the likelihood of cellular recognition leading to activation of immune and inflammatory cells. Persistent neuroinflammation is a major contributing factor to declining microelectrode recording performance. We investigated the spatial and temporal relationship of blood proteins fibrinogen and von Willebrand Factor (vWF), platelets, and type IV collagen, in relation to glial scarring markers for microglia and astrocytes following implantation of non-functional multi-shank silicon microelectrode probes into rats. Together with type IV collagen, fibrinogen and vWF augment platelet recruitment, activation, and aggregation. Our main results indicate blood proteins participating in hemostasis (fibrinogen and vWF) persisted at the microelectrode interface for up to 8-weeks after implantation. Further, type IV collagen and platelets surrounded the probe interface with similar spatial and temporal trends as vWF and fibrinogen. In addition to prolonged blood-brain barrier instability, specific blood and extracellular matrix proteins may play a role in promoting the inflammatory activation of platelets and recruitment to the microelectrode interface. STATEMENT OF SIGNIFICANCE: Implanted microelectrodes have substantial potential for restoring function to people with paralysis and amputation by providing signals that feed into natural control algorithms that drive prosthetic devices. Unfortunately, these microelectrodes do not display robust performance over time. Persistent neuroinflammation is widely thought to be a primary contributor to the devices' progressive decline in performance. Our manuscript reports on the highly local and persistent accumulation of platelets and hemostatic blood proteins around the microelectrode interface of brain implants. To our knowledge neuroinflammation driven by cellular and non-cellular responses associated with hemostasis and coagulation has not been rigorously quantified elsewhere. Our findings identify potential targets for therapeutic intervention and a better understanding of the driving mechanisms to neuroinflammation in the brain.
Collapse
Affiliation(s)
- Danny V Lam
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Anisha Javadekar
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | | | - Marina Yu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Longshun Li
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Dhariyat M Menendez
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Andrew J Shoffstall
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA.
| |
Collapse
|
23
|
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: 7] [Impact Index Per Article: 3.5] [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
|
24
|
Térémetz M, Hamdoun S, Colle F, Gerardin E, Desvilles C, Carment L, Charron S, Cuenca M, Calvet D, Baron JC, Turc G, Maier MA, Rosso C, Mas JL, Lindberg PG. Efficacy of interactive manual dexterity training after stroke: a pilot single-blinded randomized controlled trial. J Neuroeng Rehabil 2023; 20:93. [PMID: 37464404 PMCID: PMC10355015 DOI: 10.1186/s12984-023-01213-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 07/10/2023] [Indexed: 07/20/2023] Open
Abstract
OBJECTIVE To compare the efficacy of Dextrain Manipulandum™ training of dexterity components such as force control and independent finger movements, to dose-matched conventional therapy (CT) post-stroke. METHODS A prospective, single-blind, pilot randomized clinical trial was conducted. Chronic-phase post-stroke patients with mild-to-moderate dexterity impairment (Box and Block Test (BBT) > 1) received 12 sessions of Dextrain or CT. Blinded measures were obtained before and after training and at 3-months follow-up. Primary outcome was BBT-change (after-before training). Secondary outcomes included changes in motor impairments, activity limitations and dexterity components. Corticospinal excitability and short intracortical inhibition (SICI) were measured using transcranial magnetic stimulation. RESULTS BBT-change after training did not differ between the Dextrain (N = 21) vs CT group (N = 21) (median [IQR] = 5[2-7] vs 4[2-7], respectively; P = 0.36). Gains in BBT were maintained at the 3-month post-training follow-up, with a non-significant trend for enhanced BBT-change in the Dextrain group (median [IQR] = 3[- 1-7.0], P = 0.06). Several secondary outcomes showed significantly larger changes in the Dextrain group: finger tracking precision (mean ± SD = 0.3 ± 0.3N vs - 0.1 ± 0.33N; P < 0.0018), independent finger movements (34.7 ± 25.1 ms vs 7.7 ± 18.5 ms, P = 0.02) and maximal finger tapping speed (8.4 ± 7.1 vs 4.5 ± 4.9, P = 0.045). At follow-up, Dextrain group showed significantly greater improvement in Motor Activity Log (median/IQR = 0.7/0.2-0.8 vs 0.2/0.1-0.6, P = 0.05). Across both groups SICI increased in patients with greater BBT-change (Rho = 0.80, P = 0.006). Comparing Dextrain subgroups with maximal grip force higher/lower than median (61.2%), BBT-change was significantly larger in patients with low vs high grip force (7.5 ± 5.6 vs 2.9 ± 2.8; respectively, P = 0.015). CONCLUSIONS Although immediate improvements in gross dexterity post-stroke did not significantly differ between Dextrain training and CT, our findings suggest that Dextrain enhances recovery of several dexterity components and reported hand-use, particularly when motor impairment is moderate (low initial grip force). Findings need to be confirmed in a larger trial. Trial registration ClinicalTrials.gov NCT03934073 (retrospectively registered).
Collapse
Affiliation(s)
- Maxime Térémetz
- Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université Paris Cité, 102-108 Rue de La Santé, 75014, Paris, France
| | - Sonia Hamdoun
- Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université Paris Cité, 102-108 Rue de La Santé, 75014, Paris, France
- Service de Médecine Physique et de Réadaptation, Groupe Hospitalier Universitaire Paris, Psychiatrie et Neurosciences, 1 Rue Cabanis, 75014, Paris, France
| | - Florence Colle
- SSR Neurologique, Hôpitaux de Saint-Maurice, 12/14 Rue du Val d'Osne, 94410, Saint-Maurice, France
| | - Eloïse Gerardin
- Neurology Department, Stroke Unit, UCLouvain/CHU UCL Namur (Godinne), Yvoir, Belgium
| | - Claire Desvilles
- Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université Paris Cité, 102-108 Rue de La Santé, 75014, Paris, France
| | - Loïc Carment
- Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université Paris Cité, 102-108 Rue de La Santé, 75014, Paris, France
| | - Sylvain Charron
- Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université Paris Cité, 102-108 Rue de La Santé, 75014, Paris, France
| | - Macarena Cuenca
- Centre de Recherche Clinique, Groupe Hospitalier Universitaire Paris, Psychiatrie et Neurosciences, 1 Rue Cabanis, 75014, Paris, France
| | - David Calvet
- Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université Paris Cité, 102-108 Rue de La Santé, 75014, Paris, France
- Service de Neurologie, Groupe Hospitalier Universitaire Paris, Psychiatrie et Neurosciences, 1 Rue Cabanis, 75014, Paris, France
- FHU NeuroVasc, Paris, France
| | - Jean-Claude Baron
- Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université Paris Cité, 102-108 Rue de La Santé, 75014, Paris, France
- Service de Neurologie, Groupe Hospitalier Universitaire Paris, Psychiatrie et Neurosciences, 1 Rue Cabanis, 75014, Paris, France
- FHU NeuroVasc, Paris, France
| | - Guillaume Turc
- Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université Paris Cité, 102-108 Rue de La Santé, 75014, Paris, France
- Service de Neurologie, Groupe Hospitalier Universitaire Paris, Psychiatrie et Neurosciences, 1 Rue Cabanis, 75014, Paris, France
- FHU NeuroVasc, Paris, France
| | - Marc A Maier
- Université Paris Cité, CNRS, Integrative Neuroscience and Cognition Center, 75006, Paris, France
| | - Charlotte Rosso
- Institut du Cerveau et de la Moelle Épinière, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, Paris, France
| | - Jean-Louis Mas
- Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université Paris Cité, 102-108 Rue de La Santé, 75014, Paris, France
- Service de Neurologie, Groupe Hospitalier Universitaire Paris, Psychiatrie et Neurosciences, 1 Rue Cabanis, 75014, Paris, France
- FHU NeuroVasc, Paris, France
| | - Påvel G Lindberg
- Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université Paris Cité, 102-108 Rue de La Santé, 75014, Paris, France.
| |
Collapse
|
25
|
Giblin J, Kura S, Nunuez JLU, Zhang J, Kureli G, Jiang J, Boas DA, Chen IA. High throughput detection of capillary stalling events with Bessel beam two-photon microscopy. NEUROPHOTONICS 2023; 10:035009. [PMID: 37705938 PMCID: PMC10495839 DOI: 10.1117/1.nph.10.3.035009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/09/2023] [Accepted: 08/17/2023] [Indexed: 09/15/2023]
Abstract
Significance Brief disruptions in capillary flow, commonly referred to as capillary "stalling," have gained interest recently for their potential role in disrupting cerebral blood flow and oxygen delivery. Approaches to studying this phenomenon have been hindered by limited volumetric imaging rates and cumbersome manual analysis. The ability to precisely and efficiently quantify the dynamics of these events will be key in understanding their potential role in stroke and neurodegenerative diseases, such as Alzheimer's disease. Aim Our study aimed to demonstrate that the fast volumetric imaging rates offered by Bessel beam two-photon microscopy combined with improved data analysis throughput allows for faster and more precise measurement of capillary stall dynamics. Results We found that while our analysis approach was unable to achieve full automation, we were able to cut analysis time in half while also finding stalling events that were missed in traditional blind manual analysis. The resulting data showed that our Bessel beam system was captured more stalling events compared to optical coherence tomography, particularly shorter stalling events. We then compare differences in stall dynamics between a young and old group of mice as well as a demonstrate changes in stalling before and after photothrombotic model of stroke. Finally, we also demonstrate the ability to monitor arteriole dynamics alongside stall dynamics. Conclusions Bessel beam two-photon microscopy combined with high throughput analysis is a powerful tool for studying capillary stalling due to its ability to monitor hundreds of capillaries simultaneously at high frame rates.
Collapse
Affiliation(s)
- John Giblin
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Boston University, Neurophotonics Center, Boston, Massachusetts, United States
| | - Sreekanth Kura
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Boston University, Neurophotonics Center, Boston, Massachusetts, United States
| | - Juan Luis Ugarte Nunuez
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Boston University, Neurophotonics Center, Boston, Massachusetts, United States
| | - Juncheng Zhang
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Boston University, Neurophotonics Center, Boston, Massachusetts, United States
| | - Gulce Kureli
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Boston University, Neurophotonics Center, Boston, Massachusetts, United States
| | - John Jiang
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Boston University, Neurophotonics Center, Boston, Massachusetts, United States
| | - David A. Boas
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Boston University, Neurophotonics Center, Boston, Massachusetts, United States
| | - Ichun A. Chen
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Boston University, Neurophotonics Center, Boston, Massachusetts, United States
| |
Collapse
|
26
|
Sun W, Cai B, Rao J, Zhou F. Characterization of cerebrovascular changes in mice treated with alcohol by photoacoustic imaging. JOURNAL OF BIOPHOTONICS 2023:e202300038. [PMID: 37078184 DOI: 10.1002/jbio.202300038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 05/03/2023]
Abstract
Alcohol has complex effects on cerebrovascular health. Monitoring the pathology of alcohol induced cerebrovascular changes in vivo is essential for understanding the mechanism and developing potential treatment strategies. Here, photoacoustic imaging was employed to examine cerebrovascular changes in mice under the treatment of alcohol at different doses. By analyzing the association of cerebrovascular structure, hemodynamics, neuronal function and corresponding behavior, we found that alcohol affected brain function and behavior in a dose-dependent manner. Low dose of alcohol increased cerebrovascular blood volume and activated neurons, without addictive behaviors and cerebrovascular structure changes. With the dose increased, cerebrovascular blood volume gradually decreased, triggering obviously progressive effects on the immune microenvironment, cerebrovascular structure and addictive behavior. These findings will provide further insights into the characterization of the biphasic effects of alcohol.
Collapse
Affiliation(s)
- Weikang Sun
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Bingdong Cai
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Jie Rao
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Feifan Zhou
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| |
Collapse
|
27
|
Giblin JT, Park SW, Jiang J, Kılıç K, Kura S, Tang J, Boas DA, Chen IA. Measuring capillary flow dynamics using interlaced two-photon volumetric scanning. J Cereb Blood Flow Metab 2023; 43:595-609. [PMID: 36495178 PMCID: PMC10063827 DOI: 10.1177/0271678x221145091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Two photon microscopy and optical coherence tomography (OCT) are two standard methods for measuring flow speeds of red blood cells in microvessels, particularly in animal models. However, traditional two photon microscopy lacks the depth of field to adequately capture the full volumetric complexity of the cerebral microvasculature and OCT lacks the specificity offered by fluorescent labeling. In addition, the traditional raster scanning technique utilized in both modalities requires a balance of image frame rate and field of view, which severely limits the study of RBC velocities in the microvascular network. Here, we overcome this by using a custom two photon system with an axicon based Bessel beam to obtain volumetric images of the microvascular network with fluorescent specificity. We combine this with a novel scan pattern that generates pairs of frames with short time delay sufficient for tracking red blood cell flow in capillaries. We track RBC flow speeds in 10 or more capillaries simultaneously at 1 Hz in a 237 µm × 237 µm × 120 µm volume and quantified both their spatial and temporal variability in speed. We also demonstrate the ability to track flow speed changes around stalls in capillary flow and measure to 300 µm in depth.
Collapse
Affiliation(s)
- John T Giblin
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Seong-Wook Park
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - John Jiang
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Kıvılcım Kılıç
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sreekanth Kura
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Jianbo Tang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - David A Boas
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Ichun A Chen
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| |
Collapse
|
28
|
Sullender CT, Santorelli A, Richards LM, Mannava PK, Smith C, Dunn AK. Using pressure-driven flow systems to evaluate laser speckle contrast imaging. JOURNAL OF BIOMEDICAL OPTICS 2023; 28:036003. [PMID: 36915371 PMCID: PMC10007838 DOI: 10.1117/1.jbo.28.3.036003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 02/02/2023] [Indexed: 06/18/2023]
Abstract
SIGNIFICANCE Microfluidic flow phantom studies are commonly used for characterizing the performance of laser speckle contrast imaging (LSCI) instruments. The selection of the flow control system is critical for the reliable generation of flow during testing. The majority of recent LSCI studies using microfluidics used syringe pumps for flow control. AIM We quantified the uncertainty in flow generation for a syringe pump and a pressure-regulated flow system. We then assessed the performance of both LSCI and multi-exposure speckle imaging (MESI) using the pressure-regulated flow system across a range of flow speeds. APPROACH The syringe pump and pressure-regulated flow systems were evaluated during stepped flow profile experiments in a microfluidic device using an inline flow sensor. The uncertainty associated with each flow system was calculated and used to determine the reliability for instrument testing. The pressure-regulated flow system was then used to characterize the relative performance of LSCI and MESI during stepped flow profile experiments while using the inline flow sensor as reference. RESULTS The pressure-regulated flow system produced much more stable and reproducible flow outputs compared to the syringe pump. The expanded uncertainty for the syringe pump was 8 to 20 × higher than that of the pressure-regulated flow system across the tested flow speeds. Using the pressure-regulated flow system, MESI outperformed single-exposure LSCI at all flow speeds and closely mirrored the flow sensor measurements, with average errors of 4.6 % ± 2.6 % and 15.7 % ± 4.6 % , respectively. CONCLUSIONS Pressure-regulated flow systems should be used instead of syringe pumps when assessing the performance of flow measurement techniques with microfluidic studies. MESI offers more accurate relative flow measurements than traditional LSCI across a wide range of flow speeds.
Collapse
Affiliation(s)
- Colin T. Sullender
- The University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, United States
| | - Adam Santorelli
- The University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, United States
| | - Lisa M. Richards
- The University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, United States
| | - Pawan K. Mannava
- The University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, United States
| | - Christopher Smith
- The University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, United States
| | - Andrew K. Dunn
- The University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, United States
| |
Collapse
|
29
|
Smith C, Santorelli A, Engelmann S, Dunn AK. All fiber-based illumination system for multi-exposure speckle imaging. BIOMEDICAL OPTICS EXPRESS 2023; 14:771-782. [PMID: 36874493 PMCID: PMC9979660 DOI: 10.1364/boe.476178] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/15/2022] [Accepted: 12/29/2022] [Indexed: 06/18/2023]
Abstract
Monitoring blood flow is critical to treatment efficacy in many surgical settings. Laser speckle contrast imaging (LSCI) is a simple, real-time, label-free optical technique for monitoring blood flow that has emerged as a promising technique but lacks the ability to make repeatable quantitative measurements. Multi-exposure speckle imaging (MESI) is an extension of LSCI that requires increased complexity of instrumentation, which has limited its adoption. In this paper, we design and fabricate a compact, fiber-coupled MESI illumination system (FCMESI) that is substantially smaller and less complex than previous systems. Using microfluidics flow phantoms, we demonstrate that the FCMESI system measures flow with an accuracy and repeatability equivalent to traditional free space MESI illumination systems. With an in vivo stroke model, we also demonstrate the ability of FCMESI to monitor cerebral blood flow changes.
Collapse
Affiliation(s)
- Christopher Smith
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Adam Santorelli
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Shaun Engelmann
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Andrew K. Dunn
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
30
|
Chen Z, Wang X, Wu H, Fan Y, Yan Z, Lu C, Ouyang H, Zhang S, Zhang M. X-box binding protein 1 as a key modulator in “healing endothelial cells”, a novel EC phenotype promoting angiogenesis after MCAO. Cell Mol Biol Lett 2022; 27:97. [PMID: 36348288 PMCID: PMC9644469 DOI: 10.1186/s11658-022-00399-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022] Open
Abstract
Background Endothelial cells (ECs) play an important role in angiogenesis and vascular reconstruction in the pathophysiology of ischemic stroke. Previous investigations have provided a profound cerebral vascular atlas under physiological conditions, but have failed to identify new disease-related cell subtypes. We aimed to identify new EC subtypes and determine the key modulator genes. Methods Two datasets GSE174574 and GSE137482 were included in the study. Seurat was utilized as the standard quality-control pipeline. UCell was used to calculate single-cell scores to validate cellular identity. Monocle3 and CytoTRACE were utilized in aid of pseudo-time differentiation analysis. CellChat was utilized to infer the intercellular communication pathways. The angiogenesis ability of ECs was validated by MTS, Transwell, tube formation, flow cytometry, and immunofluorescence assays in vitro and in vivo. A synchrotron radiation-based propagation contrast imaging was introduced to comprehensively portray cerebral vasculature. Results We successfully identified a novel subtype of EC named “healing EC” that highly expressed pan-EC marker and pro-angiogenic genes but lowly expressed all the arteriovenous markers identified in the vascular single-cell atlas. Further analyses showed its high stemness to differentiate into other EC subtypes and potential to modulate inflammation and angiogenesis via excretion of signal molecules. We therefore identified X-box binding protein 1 (Xbp1) as a key modulator in the healing EC phenotype. In vitro and in vivo experiments confirmed its pro-angiogenic roles under both physiological and pathological conditions. Synchrotron radiation-based propagation contrast imaging further proved that Xbp1 could promote angiogenesis and recover normal vasculature conformation, especially in the corpus striatum and prefrontal cortex under middle cerebral artery occlusion (MCAO) condition. Conclusions Our study identified a novel disease-related EC subtype that showed high stemness to differentiate into other EC subtypes. The predicted molecule Xbp1 was thus confirmed as a key modulator that can promote angiogenesis and recover normal vasculature conformation. Supplementary Information The online version contains supplementary material available at 10.1186/s11658-022-00399-5.
Collapse
|
31
|
Wang X, Delle C, Asiminas A, Akther S, Vittani M, Brøgger P, Kusk P, Vo CT, Radovanovic T, Konno A, Hirai H, Fukuda M, Weikop P, Goldman SA, Nedergaard M, Hirase H. Liver-secreted fluorescent blood plasma markers enable chronic imaging of the microcirculation. CELL REPORTS METHODS 2022; 2:100302. [PMID: 36313804 PMCID: PMC9606131 DOI: 10.1016/j.crmeth.2022.100302] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 08/11/2022] [Accepted: 09/01/2022] [Indexed: 12/25/2022]
Abstract
Studying blood microcirculation is vital for gaining insights into vascular diseases. Blood flow imaging in deep tissue is currently achieved by acute administration of fluorescent dyes in the blood plasma. This is an invasive process, and the plasma fluorescence decreases within an hour of administration. Here, we report an approach for the longitudinal study of vasculature. Using a single intraperitoneal or intravenous administration of viral vectors, we express fluorescent secretory albumin-fusion proteins in the liver to chronically label the blood circulation in mice. This approach allows for longitudinal observation of circulation from 2 weeks to over 4 months after vector administration. We demonstrate the chronic assessment of vascular functions including functional hyperemia and vascular plasticity in micro- and mesoscopic scales. This genetic plasma labeling approach represents a versatile and cost-effective method for the chronic investigation of vasculature functions across the body in health and disease animal models.
Collapse
Affiliation(s)
- Xiaowen Wang
- Center for Translational Neuromedicine, Faculty of Health and Life Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christine Delle
- Center for Translational Neuromedicine, Faculty of Health and Life Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Antonis Asiminas
- Center for Translational Neuromedicine, Faculty of Health and Life Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sonam Akther
- Center for Translational Neuromedicine, Faculty of Health and Life Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marta Vittani
- Center for Translational Neuromedicine, Faculty of Health and Life Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Brøgger
- Center for Translational Neuromedicine, Faculty of Health and Life Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Kusk
- Center for Translational Neuromedicine, Faculty of Health and Life Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Camilla Trang Vo
- Center for Translational Neuromedicine, Faculty of Health and Life Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tessa Radovanovic
- Center for Translational Neuromedicine, Faculty of Health and Life Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ayumu Konno
- Viral Vector Core, Gunma University Initiative for Advanced Research, Maebashi, Gunma 371-8511, Japan
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Hirokazu Hirai
- Viral Vector Core, Gunma University Initiative for Advanced Research, Maebashi, Gunma 371-8511, Japan
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Masahiro Fukuda
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore 169857, Singapore
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Pia Weikop
- Center for Translational Neuromedicine, Faculty of Health and Life Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Steven A. Goldman
- Center for Translational Neuromedicine, Faculty of Health and Life Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Life Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Hajime Hirase
- Center for Translational Neuromedicine, Faculty of Health and Life Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
32
|
Li B, Zhao Q, Du Y, Li X, Li Z, Meng X, Li C, Meng Z, Chen J, Liu C, Cao B, Chi S. Cerebral Blood Flow Velocity Modulation and Clinical Efficacy of Acupuncture for Posterior Circulation Infarction Vertigo: A Systematic Review and Meta-Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:3740856. [PMID: 35800002 PMCID: PMC9256413 DOI: 10.1155/2022/3740856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 12/04/2022]
Abstract
Background Vertigo is a cardinal symptom of posterior circulation infarction (POCI). Acupuncture is demonstrated to have a beneficial effect on posterior circulation infarction vertigo (PCIV). However, the mechanism of acupuncture therapy is not clarified. This study aims to assess the cerebral blood flow velocity modulation and clinical efficacy of acupuncture for PCIV patients. Methods We conducted this systematic review for clinical randomized controlled trials (RCTs) regarding acupuncture on PCIV. The study duration was from September 2020 to September 2021. We searched the PubMed, EMBASE, Cochrane Library, Web of Science, Chinese Biomedical Literature Database (CBM), China National Knowledge Infrastructure (CNKI), Wanfang Database, and VIP. The publication date was set from inception to August 31, 2020. Based on the inclusion and exclusion criteria, two researchers independently screened literature and extracted data including basic study information, intervention details, outcome details, and adverse events. Outcome measures included the blood flow velocities of vertebrobasilar arteries and the Clinical Effective Rate of posterior circulation infarction vertigo. Pooled data were presented as standardized mean differences (SMDs) and relative risks (RR), with 95% confidence intervals (CIs). The meta-analysis was conducted using Review Manager software version 5.3.0. Results A total of 20 eligible RCTs (1541 participants) were included in this review, which compared acupuncture therapy (1 RCT) or acupuncture combined with pharmaceutical therapy (19 RCTs) to pharmaceutical therapy in patients with posterior circulation infarction vertigo. 7 studies assessed the blood flow velocities of the basilar artery examined by Transcranial Doppler (TCD), 8 studies assessed the bilateral vertebral arteries, and 13 studies evaluated the Clinical Effective Rate of posterior circulation infarction vertigo. Meta-analysis results showed that blood flow velocities of the basilar artery (SMD = 0.58, 95% CI = 0.40-0.76; P < 0.05), left vertebral artery (SMD = 0.48, 95% CI = 0.22-0.73; P < 0.05), and right vertebral artery (SMD = 0.44, 95% CI = 0.19-0.69; P < 0.05) were significantly higher in the acupuncture group compared with the control group. Clinical Effective Rate (RR = 1.22, 95% CI = 1.15-1.29; P = 0.792) was significantly better in the acupuncture group compared with the control group. Conclusions This study shows that acupuncture therapy is useful in improving the blood flow velocity of vertebrobasilar arteries and Clinical Effective Rate in patients with posterior circulation infarction vertigo. However, double-blind, sham-controlled trials with large sample sizes are required to support our conclusions.
Collapse
Affiliation(s)
- Boxuan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road, No. 88, Xiqing District, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Changling Road, No. 88, Xiqing District, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Beihuanan Road, Jinghai District, Tianjin, China
| | - Qi Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road, No. 88, Xiqing District, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Changling Road, No. 88, Xiqing District, Tianjin, China
| | - Yuzheng Du
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road, No. 88, Xiqing District, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Changling Road, No. 88, Xiqing District, Tianjin, China
| | - Xiayu Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road, No. 88, Xiqing District, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Changling Road, No. 88, Xiqing District, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Beihuanan Road, Jinghai District, Tianjin, China
| | - Zefang Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road, No. 88, Xiqing District, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Changling Road, No. 88, Xiqing District, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Beihuanan Road, Jinghai District, Tianjin, China
| | - Xianggang Meng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road, No. 88, Xiqing District, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Changling Road, No. 88, Xiqing District, Tianjin, China
| | - Chen Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road, No. 88, Xiqing District, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Changling Road, No. 88, Xiqing District, Tianjin, China
| | - Zhihong Meng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road, No. 88, Xiqing District, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Changling Road, No. 88, Xiqing District, Tianjin, China
| | - Junjie Chen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road, No. 88, Xiqing District, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Changling Road, No. 88, Xiqing District, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Beihuanan Road, Jinghai District, Tianjin, China
| | - Chaoda Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road, No. 88, Xiqing District, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Changling Road, No. 88, Xiqing District, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Beihuanan Road, Jinghai District, Tianjin, China
| | - Beidi Cao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road, No. 88, Xiqing District, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Changling Road, No. 88, Xiqing District, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Beihuanan Road, Jinghai District, Tianjin, China
| | - Shihao Chi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road, No. 88, Xiqing District, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Changling Road, No. 88, Xiqing District, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Beihuanan Road, Jinghai District, Tianjin, China
| |
Collapse
|
33
|
Włodarczyk L, Cichon N, Saluk-Bijak J, Bijak M, Majos A, Miller E. Neuroimaging Techniques as Potential Tools for Assessment of Angiogenesis and Neuroplasticity Processes after Stroke and Their Clinical Implications for Rehabilitation and Stroke Recovery Prognosis. J Clin Med 2022; 11:jcm11092473. [PMID: 35566599 PMCID: PMC9103133 DOI: 10.3390/jcm11092473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 02/05/2023] Open
Abstract
Stroke as the most frequent cause of disability is a challenge for the healthcare system as well as an important socio-economic issue. Therefore, there are currently a lot of studies dedicated to stroke recovery. Stroke recovery processes include angiogenesis and neuroplasticity and advances in neuroimaging techniques may provide indirect description of this action and become quantifiable indicators of these processes as well as responses to the therapeutical interventions. This means that neuroimaging and neurophysiological methods can be used as biomarkers—to make a prognosis of the course of stroke recovery and define patients with great potential of improvement after treatment. This approach is most likely to lead to novel rehabilitation strategies based on categorizing individuals for personalized treatment. In this review article, we introduce neuroimaging techniques dedicated to stroke recovery analysis with reference to angiogenesis and neuroplasticity processes. The most beneficial for personalized rehabilitation are multimodal panels of stroke recovery biomarkers, including neuroimaging and neurophysiological, genetic-molecular and clinical scales.
Collapse
Affiliation(s)
- Lidia Włodarczyk
- Department of Neurological Rehabilitation, Medical University of Lodz, Poland Milionowa 14, 93-113 Lodz, Poland
- Correspondence: (L.W.); (E.M.); Tel.: +48-(0)4-2666-77461 (E.M.); Fax: +48-(0)4-2676-1785 (E.M.)
| | - Natalia Cichon
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska, 141/143, 90-236 Lodz, Poland; (N.C.); (M.B.)
| | - Joanna Saluk-Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska, 141/143, 90-236 Lodz, Poland;
| | - Michal Bijak
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska, 141/143, 90-236 Lodz, Poland; (N.C.); (M.B.)
| | - Agata Majos
- Department of Radiological and Isotopic Diagnosis and Therapy, Medical University of Lodz, 92-213 Lodz, Poland;
| | - Elzbieta Miller
- Department of Neurological Rehabilitation, Medical University of Lodz, Poland Milionowa 14, 93-113 Lodz, Poland
- Correspondence: (L.W.); (E.M.); Tel.: +48-(0)4-2666-77461 (E.M.); Fax: +48-(0)4-2676-1785 (E.M.)
| |
Collapse
|
34
|
Myagmar BO, Chen R, Zhang X, Xu R, Jiang W, Cao W, Ji H, Zhang X. Cerebroprotein hydrolysate injection is involved in promoting long-term angiogenesis, vessel diameter and density after cerebral ischemia in mice. Life Sci 2022; 300:120568. [PMID: 35489566 DOI: 10.1016/j.lfs.2022.120568] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 12/09/2022]
Abstract
AIMS In this study, we aimed investigate the impacts of CH-I on angiogenesis, effects for vascular structure changes and long-term neurological recovery after ischemic stroke as well as the potential mechanisms. MAIN METHODS Young male mice subjected to intraluminal middle cerebral artery occlusion were administrated with CH-I once daily from day 1 to day 14 after stroke. The infarct volume was evaluated by TTC staining at day 7 after stroke. Neurological deficits were measured 1 to 28 days after stroke. Microvascular density, astrocyte coverage, and angiogenesis were assessed by IF, qRT-PCR, and WB at regular intervals after stroke. LSCI and TPMI measured changes in blood flow and vascular density and width from the day after stroke to day 28. KEY FINDINGS Compared with the dMCAO group, CH-I treatment significantly improved neurological recovery and reduced the infarct at day 7 after stroke. CH-I treatment increased the expression of the CD31, BrdU+/CD31+ microvessels and GFAP positive vessels in the peri-infarct cortex at day 7 to 28 after stroke. The expression of protein and gene were enhanced in CH-I group. CH-I significantly improved cerebral blood flow at day 7 after stroke. CH-I increased the vascular density and vascular width at day 14 after stroke. SIGNIFICANCE CH-I has been shown to restore nerve function, reduce the rate of cerebral infarction, increase microvascular density, and promote angiogenesis. CH-I improved cerebral blood flow, protected blood vessels from postoperative stenosis, and improved vascular plasticity.
Collapse
Affiliation(s)
- Bat-Otgon Myagmar
- Department of Neurology, Second Hospital of Hebei Medical University, Hebei Medical University Shijiazhuang, Hebei 050000, People's Republic of China
| | - Rong Chen
- Hebei Collaborative Innovation Center for Cardio- Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Xiao Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Hebei Medical University Shijiazhuang, Hebei 050000, People's Republic of China
| | - Renhao Xu
- Hebei Collaborative Innovation Center for Cardio- Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Wei Jiang
- Hebei Collaborative Innovation Center for Cardio- Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Wen Cao
- Department of Neurology, Second Hospital of Hebei Medical University, Hebei Medical University Shijiazhuang, Hebei 050000, People's Republic of China
| | - Hui Ji
- Department of Neurology, Second Hospital of Hebei Medical University, Hebei Medical University Shijiazhuang, Hebei 050000, People's Republic of China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Hebei Medical University Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Collaborative Innovation Center for Cardio- Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China.
| |
Collapse
|
35
|
Lin X, Chen L, Jullienne A, Zhang H, Salehi A, Hamer M, C. Holmes T, Obenaus A, Xu X. Longitudinal dynamics of microvascular recovery after acquired cortical injury. Acta Neuropathol Commun 2022; 10:59. [PMID: 35468870 PMCID: PMC9036719 DOI: 10.1186/s40478-022-01361-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/05/2022] [Indexed: 01/04/2023] Open
Abstract
Acquired brain injuries due to trauma damage the cortical vasculature, which in turn impairs blood flow to injured tissues. There are reports of vascular morphological recovery following traumatic brain injury, but the remodeling process has not been examined longitudinally in detail after injury in vivo. Understanding the dynamic processes that influence recovery is thus critically important. We evaluated the longitudinal and dynamic microvascular recovery and remodeling up to 2 months post injury using live brain miniscope and 2-photon microscopic imaging. The new imaging approaches captured dynamic morphological and functional recovery processes at high spatial and temporal resolution in vivo. Vessel painting documented the initial loss and subsequent temporal morphological vascular recovery at the injury site. Miniscopes were used to longitudinally image the temporal dynamics of vascular repair in vivo after brain injury in individual mice across each cohort. We observe near-immediate nascent growth of new vessels in and adjacent to the injury site that peaks between 14 and 21 days post injury. 2-photon microscopy confirms new vascular growth and further demonstrates differences between cortical layers after cortical injury: large vessels persist in deeper cortical layers (> 200 μm), while superficial layers exhibit a dense plexus of fine (and often non-perfused) vessels displaying regrowth. Functionally, blood flow increases mirror increasing vascular density. Filopodia development and endothelial sprouting is measurable within 3 days post injury that rapidly transforms regions devoid of vessels to dense vascular plexus in which new vessels become increasingly perfused. Within 7 days post injury, blood flow is observed in these nascent vessels. Behavioral analysis reveals improved vascular modulation after 9 days post injury, consistent with vascular regrowth. We conclude that morphological recovery events are closely linked to functional recovery of blood flow to the compromised tissues, which subsequently leads to improved behavioral outcomes.
Collapse
|
36
|
Longo V, Barbati SA, Re A, Paciello F, Bolla M, Rinaudo M, Miraglia F, Alù F, Di Donna MG, Vecchio F, Rossini PM, Podda MV, Grassi C. Transcranial Direct Current Stimulation Enhances Neuroplasticity and Accelerates Motor Recovery in a Stroke Mouse Model. Stroke 2022; 53:1746-1758. [PMID: 35291824 DOI: 10.1161/strokeaha.121.034200] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND More effective strategies are needed to promote poststroke functional recovery. Here, we evaluated the impact of bihemispheric transcranial direct current stimulation (tDCS) on forelimb motor function recovery and the underlying mechanisms in mice subjected to focal ischemia of the motor cortex. METHODS Photothrombotic stroke was induced in the forelimb brain motor area, and tDCS was applied once per day for 3 consecutive days, starting 72 hours after stroke. Grid-walking, single pellet reaching, and grip strength tests were conducted to assess motor function. Local field potentials were recorded to evaluate brain connectivity. Western immunoblotting, ELISA, quantitative real-time polymerase chain reaction, and Golgi-Cox staining were used to uncover tDCS-mediated stroke recovery mechanisms. RESULTS Among our results, tDCS increased the rate of motor recovery, anticipating it at the early subacute stage. In this window, tDCS enhanced BDNF (brain-derived neurotrophic factor) expression and dendritic spine density in the peri-infarct motor cortex, along with increasing functional connectivity between motor and somatosensory cortices. Treatment with the BDNF TrkB (tropomyosin-related tyrosine kinase B) receptor inhibitor, ANA-12, prevented tDCS effects on motor recovery and connectivity as well as the increase of spine density, pERK (phosphorylated extracellular signal-regulated kinase), pCaMKII (phosphorylated calcium/calmodulin-dependent protein kinase II), pMEF (phosphorylated myocyte-enhancer factor), and PSD (postsynaptic density)-95. The tDCS-promoted rescue was paralleled by enhanced plasma BDNF level, suggesting its potential role as circulating prognostic biomarker. CONCLUSIONS The rate of motor recovery is accelerated by tDCS applied in the subacute phase of stroke. Anticipation of motor recovery via vicariate pathways or neural reserve recruitment would potentially enhance the efficacy of standard treatments, such as physical therapy, which is often delayed to a later stage when plastic responses are progressively lower.
Collapse
Affiliation(s)
- Valentina Longo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy (V.L., S.A.B., A.R., F.P., M.B., M.R., M.G.D.D., M.V.P., C.G.)
| | - Saviana Antonella Barbati
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy (V.L., S.A.B., A.R., F.P., M.B., M.R., M.G.D.D., M.V.P., C.G.)
| | - Agnese Re
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy (V.L., S.A.B., A.R., F.P., M.B., M.R., M.G.D.D., M.V.P., C.G.)
| | - Fabiola Paciello
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy (V.L., S.A.B., A.R., F.P., M.B., M.R., M.G.D.D., M.V.P., C.G.)
| | - Maria Bolla
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy (V.L., S.A.B., A.R., F.P., M.B., M.R., M.G.D.D., M.V.P., C.G.)
| | - Marco Rinaudo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy (V.L., S.A.B., A.R., F.P., M.B., M.R., M.G.D.D., M.V.P., C.G.)
| | - Francesca Miraglia
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele Roma, Italy (F.M., F.A., F.V., P.M.R.)
| | - Francesca Alù
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele Roma, Italy (F.M., F.A., F.V., P.M.R.)
| | - Martina Gaia Di Donna
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy (V.L., S.A.B., A.R., F.P., M.B., M.R., M.G.D.D., M.V.P., C.G.)
| | - Fabrizio Vecchio
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele Roma, Italy (F.M., F.A., F.V., P.M.R.).,eCampus University, Novedrate, Como, Italy (F.V.)
| | - Paolo Maria Rossini
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele Roma, Italy (F.M., F.A., F.V., P.M.R.)
| | - Maria Vittoria Podda
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy (V.L., S.A.B., A.R., F.P., M.B., M.R., M.G.D.D., M.V.P., C.G.).,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (M.V.P., C.G.)
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy (V.L., S.A.B., A.R., F.P., M.B., M.R., M.G.D.D., M.V.P., C.G.).,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy (M.V.P., C.G.)
| |
Collapse
|
37
|
Krzyspiak J, Yan J, Ghosh HS, Galinski B, Lituma PJ, Alvina K, Quezada A, Kee S, Grońska-Pęski M, Tai YD, McDermott K, Gonçalves JT, Zukin RS, Weiser DA, Castillo PE, Khodakhah K, Hébert JM. Donor-derived vasculature is required to support neocortical cell grafts after stroke. Stem Cell Res 2022; 59:102642. [PMID: 34971934 PMCID: PMC11495404 DOI: 10.1016/j.scr.2021.102642] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 12/14/2021] [Accepted: 12/23/2021] [Indexed: 11/27/2022] Open
Abstract
Neural precursor cells (NPCs) transplanted into the adult neocortex generate neurons that synaptically integrate with host neurons, supporting the possibility of achieving functional tissue repair. However, poor survival and functional neuronal recovery of transplanted NPCs greatly limits engraftment. Here, we test the hypothesis that combining blood vessel-forming vascular cells with neuronal precursors improves engraftment. By transplanting mixed embryonic neocortical cells into adult mice with neocortical strokes, we show that transplant-derived neurons synapse with appropriate targets while donor vascular cells form vessels that fuse with the host vasculature to perfuse blood within the graft. Although all grafts became vascularized, larger grafts had greater contributions of donor-derived vessels that increased as a function of their distance from the host-graft border. Moreover, excluding vascular cells from the donor cell population strictly limited graft size. Thus, inclusion of vessel-forming vascular cells with NPCs is required for more efficient engraftment and ultimately for tissue repair.
Collapse
Affiliation(s)
- Joanna Krzyspiak
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Stem Cell Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jingqi Yan
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hiyaa S Ghosh
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Basia Galinski
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Pablo J Lituma
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Karina Alvina
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alexandra Quezada
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Stem Cell Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Samantha Kee
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Marta Grońska-Pęski
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Stem Cell Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yi De Tai
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; University of Rochester, Rochester, NY, USA
| | - Kelsey McDermott
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - J Tiago Gonçalves
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Stem Cell Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - R Suzanne Zukin
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Daniel A Weiser
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Pablo E Castillo
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kamran Khodakhah
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jean M Hébert
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Stem Cell Institute, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
38
|
Rohden F, Teixeira LV, Bernardi LP, Ferreira PCL, Colombo M, Teixeira GR, de Oliveira FDS, Cirne Lima EO, Guma FCR, Souza DO. Functional Recovery Caused by Human Adipose Tissue Mesenchymal Stem Cell-Derived Extracellular Vesicles Administered 24 h after Stroke in Rats. Int J Mol Sci 2021; 22:12860. [PMID: 34884665 PMCID: PMC8657917 DOI: 10.3390/ijms222312860] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 12/11/2022] Open
Abstract
Ischemic stroke is a major cause of death and disability, intensely demanding innovative and accessible therapeutic strategies. Approaches presenting a prolonged period for therapeutic intervention and new treatment administration routes are promising tools for stroke treatment. Here, we evaluated the potential neuroprotective properties of nasally administered human adipose tissue mesenchymal stem cell (hAT-MSC)-derived extracellular vesicles (EVs) obtained from healthy individuals who underwent liposuction. After a single intranasal EV (200 µg/kg) administered 24 h after a focal permanent ischemic stroke in rats, a higher number of EVs, improvement of the blood-brain barrier, and re-stabilization of vascularization were observed in the recoverable peri-infarct zone, as well as a significant decrease in infarct volume. In addition, EV treatment recovered long-term motor (front paws symmetry) and behavioral impairment (short- and long-term memory and anxiety-like behavior) induced by ischemic stroke. In line with these findings, our work highlights hAT-MSC-derived EVs as a promising therapeutic strategy for stroke.
Collapse
Affiliation(s)
- Francieli Rohden
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
- Instituto de Cardiologia do Rio Grande do Sul Fundação Universitária de Cardiologia, Porto Alegre 90620-101, Brazil
| | - Luciele Varaschini Teixeira
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
- Instituto de Cardiologia do Rio Grande do Sul Fundação Universitária de Cardiologia, Porto Alegre 90620-101, Brazil
| | - Luis Pedro Bernardi
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
- Faculty of Biomedicine, Universidade Federal de Ciências da Saúde de Porto Alegre—UFCSPA, Porto Alegre 90050-170, Brazil
| | - Pamela Cristina Lukasewicz Ferreira
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
| | - Mariana Colombo
- Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil;
| | - Geciele Rodrigues Teixeira
- Experimental Research Center, Reproductive and Cellular Pharmacology Laboratory, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, Brazil; (G.R.T.); (F.d.S.d.O.); (E.O.C.L.)
| | - Fernanda dos Santos de Oliveira
- Experimental Research Center, Reproductive and Cellular Pharmacology Laboratory, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, Brazil; (G.R.T.); (F.d.S.d.O.); (E.O.C.L.)
| | - Elizabeth Obino Cirne Lima
- Experimental Research Center, Reproductive and Cellular Pharmacology Laboratory, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, Brazil; (G.R.T.); (F.d.S.d.O.); (E.O.C.L.)
| | - Fátima Costa Rodrigues Guma
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
| | - Diogo Onofre Souza
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul—UFRGS, Porto Alegre 90040-60, Brazil; (L.V.T.); (L.P.B.); (P.C.L.F.); (F.C.R.G.)
| |
Collapse
|
39
|
Zhang Q, Gheres KW, Drew PJ. Origins of 1/f-like tissue oxygenation fluctuations in the murine cortex. PLoS Biol 2021; 19:e3001298. [PMID: 34264930 PMCID: PMC8282088 DOI: 10.1371/journal.pbio.3001298] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 05/24/2021] [Indexed: 01/07/2023] Open
Abstract
The concentration of oxygen in the brain spontaneously fluctuates, and the distribution of power in these fluctuations has a 1/f-like spectra, where the power present at low frequencies of the power spectrum is orders of magnitude higher than at higher frequencies. Though these oscillations have been interpreted as being driven by neural activity, the origin of these 1/f-like oscillations is not well understood. Here, to gain insight of the origin of the 1/f-like oxygen fluctuations, we investigated the dynamics of tissue oxygenation and neural activity in awake behaving mice. We found that oxygen signal recorded from the cortex of mice had 1/f-like spectra. However, band-limited power in the local field potential did not show corresponding 1/f-like fluctuations. When local neural activity was suppressed, the 1/f-like fluctuations in oxygen concentration persisted. Two-photon measurements of erythrocyte spacing fluctuations and mathematical modeling show that stochastic fluctuations in erythrocyte flow could underlie 1/f-like dynamics in oxygenation. These results suggest that the discrete nature of erythrocytes and their irregular flow, rather than fluctuations in neural activity, could drive 1/f-like fluctuations in tissue oxygenation.
Collapse
Affiliation(s)
- Qingguang Zhang
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail: (QZ); (PJD)
| | - Kyle W. Gheres
- Graduate Program in Molecular Cellular and Integrative Biosciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Patrick J. Drew
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Neurosurgery, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail: (QZ); (PJD)
| |
Collapse
|
40
|
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: 97] [Impact Index Per Article: 24.3] [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
|