51
|
Khan J, Rudrapal M, Bhat EA, Ali A, Alaidarous M, Alshehri B, Banwas S, Ismail R, Egbuna C. Perspective Insights to Bio-Nanomaterials for the Treatment of Neurological Disorders. Front Bioeng Biotechnol 2021; 9:724158. [PMID: 34712651 PMCID: PMC8546296 DOI: 10.3389/fbioe.2021.724158] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/20/2021] [Indexed: 12/25/2022] Open
Abstract
The significance of biomaterials is well appreciated in nanotechnology, and its use has resulted in major advances in biomedical sciences. Although, currently, very little data is available on the clinical trial studies for treatment of neurological conditions, numerous promising advancements have been reported in drug delivery and regenerative therapies which can be applied in clinical practice. Among the commonly reported biomaterials in literature, the self-assembling peptides and hydrogels have been recognized as the most potential candidate for treatment of common neurological conditions such as Alzheimer's, Parkinson's, spinal cord injury, stroke and tumors. The hydrogels, specifically, offer advantages like flexibility and porosity, and mimics the properties of the extracellular matrix of the central nervous system. These factors make them an ideal scaffold for drug delivery through the blood-brain barrier and tissue regeneration (using stem cells). Thus, the use of biomaterials as suitable matrix for therapeutic purposes has emerged as a promising area of neurosciences. In this review, we describe the application of biomaterials, and the current advances, in treatment of statistically common neurological disorders.
Collapse
Affiliation(s)
- Johra Khan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia
- Health and Basic Sciences Research Center, Majmaah University, Majmaah, Saudi Arabia
| | - Mithun Rudrapal
- Rasiklal M. Dhariwal Institute of Pharmaceutical Education & Research, Pune, India
| | - Eijaz Ahmed Bhat
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, India
| | - Ahmad Ali
- Department of Life Sciences, University of Mumbai, Mumbai, India
| | - Mohammad Alaidarous
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia
- Health and Basic Sciences Research Center, Majmaah University, Majmaah, Saudi Arabia
| | - Bader Alshehri
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia
- Health and Basic Sciences Research Center, Majmaah University, Majmaah, Saudi Arabia
| | - Saeed Banwas
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia
- Health and Basic Sciences Research Center, Majmaah University, Majmaah, Saudi Arabia
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR, United States
| | - Randa Ismail
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia
- Health and Basic Sciences Research Center, Majmaah University, Majmaah, Saudi Arabia
| | - Chukwuebuka Egbuna
- World Bank Africa Centre of Excellence in Public Health and Toxicological Research (PUTOR), University of Port Harcourt, Port Harcourt, Nigeria
- Department of Biochemistry, University of Port Harcourt, Port Harcourt, Nigeria
| |
Collapse
|
52
|
Bahlakeh G, Rahbarghazi R, Mohammadnejad D, Abedelahi A, Karimipour M. Current knowledge and challenges associated with targeted delivery of neurotrophic factors into the central nervous system: focus on available approaches. Cell Biosci 2021; 11:181. [PMID: 34641969 PMCID: PMC8507154 DOI: 10.1186/s13578-021-00694-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/28/2021] [Indexed: 12/23/2022] Open
Abstract
During the last decades, numerous basic and clinical studies have been conducted to assess the delivery efficiency of therapeutic agents into the brain and spinal cord parenchyma using several administration routes. Among conventional and in-progress administrative routes, the eligibility of stem cells, viral vectors, and biomaterial systems have been shown in the delivery of NTFs. Despite these manifold advances, the close association between the delivery system and regeneration outcome remains unclear. Herein, we aimed to discuss recent progress in the delivery of these factors and the pros and cons related to each modality.
Collapse
Affiliation(s)
- Gozal Bahlakeh
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Daruosh Mohammadnejad
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Abedelahi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Karimipour
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. .,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
53
|
Lamtahri R, Hazime M, Gowing EK, Nagaraja RY, Maucotel J, Alasoadura M, Quilichini PP, Lehongre K, Lefranc B, Gach-Janczak K, Marcher AB, Mandrup S, Vaudry D, Clarkson AN, Leprince J, Chuquet J. The Gliopeptide ODN, a Ligand for the Benzodiazepine Site of GABA A Receptors, Boosts Functional Recovery after Stroke. J Neurosci 2021; 41:7148-7159. [PMID: 34210784 PMCID: PMC8372017 DOI: 10.1523/jneurosci.2255-20.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/14/2020] [Accepted: 03/25/2021] [Indexed: 11/21/2022] Open
Abstract
Following stroke, the survival of neurons and their ability to reestablish connections is critical to functional recovery. This is strongly influenced by the balance between neuronal excitation and inhibition. In the acute phase of experimental stroke, lethal hyperexcitability can be attenuated by positive allosteric modulation of GABAA receptors (GABAARs). Conversely, in the late phase, negative allosteric modulation of GABAAR can correct the suboptimal excitability and improves both sensory and motor recovery. Here, we hypothesized that octadecaneuropeptide (ODN), an endogenous allosteric modulator of the GABAAR synthesized by astrocytes, influences the outcome of ischemic brain tissue and subsequent functional recovery. We show that ODN boosts the excitability of cortical neurons, which makes it deleterious in the acute phase of stroke. However, if delivered after day 3, ODN is safe and improves motor recovery over the following month in two different paradigms of experimental stroke in mice. Furthermore, we bring evidence that, during the subacute period after stroke, the repairing cortex can be treated with ODN by means of a single hydrogel deposit into the stroke cavity.SIGNIFICANCE STATEMENT Stroke remains a devastating clinical challenge because there is no efficient therapy to either minimize neuronal death with neuroprotective drugs or to enhance spontaneous recovery with neurorepair drugs. Around the brain damage, the peri-infarct cortex can be viewed as a reservoir of plasticity. However, the potential of wiring new circuits in these areas is restrained by a chronic excess of GABAergic inhibition. Here we show that an astrocyte-derived peptide, can be used as a delayed treatment, to safely correct cortical excitability and facilitate sensorimotor recovery after stroke.
Collapse
Affiliation(s)
- Rhita Lamtahri
- Normandie Université, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1239, Neuronal and Neuroendocrine Differentiation and Communication, Rouen, France
| | - Mahmoud Hazime
- Normandie Université, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1239, Neuronal and Neuroendocrine Differentiation and Communication, Rouen, France
| | - Emma K Gowing
- Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, 76000, 9054, New Zealand
| | - Raghavendra Y Nagaraja
- Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, 76000, 9054, New Zealand
| | - Julie Maucotel
- Normandie Université, UNIROUEN, Animal Facility, Rouen, 76000, France
| | - Michael Alasoadura
- Normandie Université, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1239, Neuronal and Neuroendocrine Differentiation and Communication, Rouen, France
| | | | - Katia Lehongre
- Inserm U 1127, Centre National de la Recherche Scientifique Unite Mixte de Recherche 7225, Sorbonne Universités, UPMC Univ Paris 06 Unite Mixte de Recherche S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France
| | - Benjamin Lefranc
- Normandie Université, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1239, Neuronal and Neuroendocrine Differentiation and Communication, Rouen, France
- Institute for Research and Innovation in Biomedicine, Normandie Université, PRIMACEN, Rouen, 76000, France
| | - Katarzyna Gach-Janczak
- Normandie Université, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1239, Neuronal and Neuroendocrine Differentiation and Communication, Rouen, France
- Department of Biomolecular Chemistry, Medicinal University of Łódź, Łódź, 90-137, Poland
| | - Ann-Britt Marcher
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, 5230, Denmark
| | - Susanne Mandrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, 5230, Denmark
| | - David Vaudry
- Normandie Université, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1239, Neuronal and Neuroendocrine Differentiation and Communication, Rouen, France
- Institute for Research and Innovation in Biomedicine, Normandie Université, PRIMACEN, Rouen, 76000, France
| | - Andrew N Clarkson
- Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, 76000, 9054, New Zealand
| | - Jérôme Leprince
- Normandie Université, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1239, Neuronal and Neuroendocrine Differentiation and Communication, Rouen, France
- Institute for Research and Innovation in Biomedicine, Normandie Université, PRIMACEN, Rouen, 76000, France
| | - Julien Chuquet
- Normandie Université, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1239, Neuronal and Neuroendocrine Differentiation and Communication, Rouen, France
| |
Collapse
|
54
|
Thomas JM, Louca I, Bolan F, Sava O, Allan SM, Lawrence CB, Pinteaux E. Regenerative Potential of Hydrogels for Intracerebral Hemorrhage: Lessons from Ischemic Stroke and Traumatic Brain Injury Research. Adv Healthc Mater 2021; 10:e2100455. [PMID: 34197036 PMCID: PMC11468990 DOI: 10.1002/adhm.202100455] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/15/2021] [Indexed: 01/02/2023]
Abstract
Intracerebral hemorrhage (ICH) is a deadly and debilitating type of stroke, caused by the rupture of cerebral blood vessels. To date, there are no restorative interventions approved for use in ICH patients, highlighting a critical unmet need. ICH shares some pathological features with other acute brain injuries such as ischemic stroke (IS) and traumatic brain injury (TBI), including the loss of brain tissue, disruption of the blood-brain barrier, and activation of a potent inflammatory response. New biomaterials such as hydrogels have been recently investigated for their therapeutic benefit in both experimental IS and TBI, owing to their provision of architectural support for damaged brain tissue and ability to deliver cellular and molecular therapies. Conversely, research on the use of hydrogels for ICH therapy is still in its infancy, with very few published reports investigating their therapeutic potential. Here, the published use of hydrogels in experimental ICH is commented upon and how approaches reported in the IS and TBI fields may be applied to ICH research to inform the design of future therapies is described. Unique aspects of ICH that are distinct from IS and TBI that should be considered when translating biomaterial-based therapies between disease models are also highlighted.
Collapse
Affiliation(s)
- Josephine M. Thomas
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Irene Louca
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Faye Bolan
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Oana‐Roxana Sava
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Stuart M. Allan
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Catherine B. Lawrence
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Emmanuel Pinteaux
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| |
Collapse
|
55
|
Ngo MT, Harley BAC. Progress in mimicking brain microenvironments to understand and treat neurological disorders. APL Bioeng 2021; 5:020902. [PMID: 33869984 PMCID: PMC8034983 DOI: 10.1063/5.0043338] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/22/2021] [Indexed: 12/16/2022] Open
Abstract
Neurological disorders including traumatic brain injury, stroke, primary and metastatic brain tumors, and neurodegenerative diseases affect millions of people worldwide. Disease progression is accompanied by changes in the brain microenvironment, but how these shifts in biochemical, biophysical, and cellular properties contribute to repair outcomes or continued degeneration is largely unknown. Tissue engineering approaches can be used to develop in vitro models to understand how the brain microenvironment contributes to pathophysiological processes linked to neurological disorders and may also offer constructs that promote healing and regeneration in vivo. In this Perspective, we summarize features of the brain microenvironment in normal and pathophysiological states and highlight strategies to mimic this environment to model disease, investigate neural stem cell biology, and promote regenerative healing. We discuss current limitations and resulting opportunities to develop tissue engineering tools that more faithfully recapitulate the aspects of the brain microenvironment for both in vitro and in vivo applications.
Collapse
Affiliation(s)
- Mai T. Ngo
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Brendan A. C. Harley
- Author to whom correspondence should be addressed:. Tel.: (217) 244-7112. Fax: (217) 333-5052
| |
Collapse
|
56
|
Ucar B. Natural biomaterials in brain repair: A focus on collagen. Neurochem Int 2021; 146:105033. [PMID: 33785419 DOI: 10.1016/j.neuint.2021.105033] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 03/07/2021] [Accepted: 03/22/2021] [Indexed: 12/16/2022]
Abstract
Biomaterials derived from natural resources have increasingly been used for versatile applications in the central nervous system (CNS). Thanks to their biocompatibility and biodegradability, natural biomaterials offer vast possibilities for future clinical repair strategies for the CNS. These materials can be used for diverse applications such as hydrogels to fill the tissue cavities, microparticles to deliver drugs across the blood-brain barrier, and scaffolds to transplant stem cells. In this review, various uses of prominent protein and polysaccharide biomaterials, with a special focus on collagen, in repair and regenerative applications for the brain are summarized together with their individual advantages and disadvantages.
Collapse
Affiliation(s)
- Buket Ucar
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, Austria.
| |
Collapse
|
57
|
Zhang X, Chen F, Wang Y. Commentary: In vivo Neuroregeneration to Treat Ischemic Stroke Through NeuroD1 AAV-Based Gene Therapy in Adult Non-human Primates. Front Cell Dev Biol 2021; 9:648020. [PMID: 34124038 PMCID: PMC8194073 DOI: 10.3389/fcell.2021.648020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/12/2021] [Indexed: 01/21/2023] Open
Affiliation(s)
- Xiaoqin Zhang
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| | - Fenghua Chen
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| | - Youcui Wang
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| |
Collapse
|
58
|
DeBoer SR, Hubbard R, Mersha M, Pinilla Monsalve G, Winter S, Zeiler SR. Enhanced Spontaneous Motor Recovery After Stroke in Mice Treated With Cerebrolysin. Neurorehabil Neural Repair 2021; 35:525-533. [PMID: 33955296 DOI: 10.1177/15459683211000734] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Motor recovery after stroke in humans and in rodent models is time sensitive. Recovery in patients is a result of biological spontaneous recovery via endogenous repair mechanisms and is likely improved by enhancing the synaptic plasticity required for endogenous repair. Cerebrolysin is a polypeptide preparation known to enhance neuroplasticity and may improve recovery in patients. In mice, we tested the hypothesis that Cerebrolysin can act poststroke to enhance both spontaneous and training-associated motor recovery. METHODS Mice were trained to perform a skilled prehension task. We then induced a photothrombotic stroke in the caudal forelimb area, after which we retrained animals on the prehension task in the presence or absence of Cerebrolysin after a 2-day or 8-day delay. Mice received daily intraperitoneal Cerebrolysin or saline injections starting poststroke day 1 or poststroke day 7. RESULTS Prior studies showed that poststroke recovery of prehension can occur if animals receive rehabilitative training during an early sensitive period but is incomplete if rehabilitative training is delayed. In contrast, we show complete recovery of prehension, despite a delay in rehabilitative training, when mice receive daily Cerebrolysin administration starting on poststroke day 1 or on poststroke day 8. When Cerebrolysin is given on poststroke day 1, recovery occurred even in the absence of training. Stroke volumes were similar across groups. CONCLUSIONS Poststroke Cerebrolysin administration leads to recovery of motor function independent of rehabilitative training without a protective effect on stroke volume. This is one of the first demonstrations of training-independent motor recovery in rodent stroke models.
Collapse
Affiliation(s)
| | | | | | | | - Stefan Winter
- Ever Neuro Pharma GmbH, Unterach, Oberösterreich, Austria
| | | |
Collapse
|
59
|
Ali MA, Bhuiyan MH. Types of biomaterials useful in brain repair. Neurochem Int 2021; 146:105034. [PMID: 33789130 DOI: 10.1016/j.neuint.2021.105034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/28/2021] [Accepted: 03/22/2021] [Indexed: 01/21/2023]
Abstract
Biomaterials is an emerging field in the study of brain tissue engineering and repair or neurogenesis. The fabrication of biomaterials that can replicate the mechanical and viscoelastic features required by the brain, including the poroviscoelastic responses, force dissipation, and solute diffusivity are essential to be mapped from the macro to the nanoscale level under physiological conditions in order for us to gain an effective treatment for neurodegenerative diseases. This research topic has identified a critical study gap that must be addressed, and that is to source suitable biomaterials and/or create reliable brain-tissue-like biomaterials. This chapter will define and discuss the various types of biomaterials, their structures, and their function-properties features which would enable the development of next-generation biomaterials useful in brain repair.
Collapse
Affiliation(s)
- M Azam Ali
- Center for Bioengineering and Nanomedicine, Faculty of Dentistry, Division of Health Sciences, University of Otago, Dunedin, New Zealand.
| | - Mozammel Haque Bhuiyan
- Center for Bioengineering and Nanomedicine, Faculty of Dentistry, Division of Health Sciences, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
60
|
Lin Y, Yao M, Wu H, Wu F, Cao S, Ni H, Dong J, Yang D, Sun Y, Kou X, Li J, Xiao H, Chang L, Wu J, Liu Y, Luo C, Zhu D. Environmental enrichment implies GAT-1 as a potential therapeutic target for stroke recovery. Theranostics 2021; 11:3760-3780. [PMID: 33664860 PMCID: PMC7914370 DOI: 10.7150/thno.53316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/06/2021] [Indexed: 12/21/2022] Open
Abstract
Rationale: Stroke is a leading cause of adult disability worldwide, but no drug provides functional recovery during the repair phase. Accumulating evidence demonstrates that environmental enrichment (EE) promotes stroke recovery by enhancing network excitability. However, the complexities of utilizing EE in a clinical setting limit its translation. Methods: We used multifaceted approaches combining electrophysiology, chemogenetics, optogenetics, and floxed mice in a mouse photothrombotic stroke model to reveal the key target of EE-mediated stroke recovery. Results: EE reduced tonic gamma-aminobutyric acid (GABA) inhibition and facilitated phasic GABA inhibition in the peri-infarct cortex, thereby promoting network excitability and stroke recovery. These beneficial effects depended on GAT-1, a GABA transporter regulating both tonic and phasic GABA signaling, as EE positively regulated GAT-1 expression, trafficking, and function. Furthermore, GAT-1 was necessary for EE-induced network plasticity, including structural neuroplasticity, input synaptic strengthening in the peri-infarct cortex, output synaptic strengthening in the corticospinal tract, and sprouting of uninjured corticospinal axons across the midline into the territory of denervated spinal cord, and functional recovery from stroke. Moreover, restoration of GAT-1 function in the peri-infarct cortex by its overexpression showed similar beneficial effects on stroke recovery as EE exposure. Conclusion: GAT-1 is a key molecular substrate of the effects of EE on network excitability and consequent stroke recovery and can serve as a novel therapeutic target for stroke treatment during the repair phase.
Collapse
|
61
|
Glickman RD, Onorato M, Campos MM, O'Boyle MP, Singh RK, Zarembinski TI, Binette F, Nasonkin IO. Intraocular Injection of HyStem Hydrogel Is Tolerated Well in the Rabbit Eye. J Ocul Pharmacol Ther 2021; 37:60-71. [PMID: 33449859 DOI: 10.1089/jop.2020.0042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Purpose: To determine the long-term biocompatibility of HyStem® hydrogel in the rabbit eye for use as a carrier for cell or drug delivery into the ocular space. Methods: HyStem hydrogel formulation solidifies ∼20 min after reconstitution, thus can potentially form a solid deposit after injection in situ. To study the ocular disposition of fluorescein-labeled HyStem, we delivered 50 μL/eye over 1 min into the vitreous space of the rabbit. We used 3 Dutch-Belted and 3 New Zealand-pigmented rabbits, all females, delivered the gel into the right eyes, and injected 50 μL BSS Plus into the left eyes as a control. Retinal morphology was assessed by optical coherence tomography (OCT) and white light fundus photography. Fluorescence fundus photography enabled measurement of the clearance of the labeled hydrogel from the posterior chamber. Visual function was evaluated using flash and flicker electroretinography (ERG) pre- and postinjection and at weekly intervals thereafter for 6 weeks. Retinal immunohistochemistry for microglial inflammatory markers was carried out with antiglial fibrillary acidic protein (GFAP) antibody, isolectin B4 (IB4), and 4',6-diamidino-2-phenylindole (DAPI). Results: The gel was successfully delivered into the vitreous space without the formation of a discrete retinal deposit. Fundus imaging, OCT measurements of retinal thickness, and immunohistochemical data indicated an absence of retinal inflammation, and ERG indicated no impact on retinal function. The half-time of HyStem clearance calculated from the loss of fundus fluorescence was 3.9 days. Conclusions: HyStem hydrogel appears to be biocompatible in the ocular space of a large eye and safe for long-term intraocular application.
Collapse
Affiliation(s)
- Randolph D Glickman
- Department of Ophthalmology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | | | - Maria M Campos
- Histopathology Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael P O'Boyle
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | | | | | | | | |
Collapse
|
62
|
Bressan C, Saghatelyan A. Intrinsic Mechanisms Regulating Neuronal Migration in the Postnatal Brain. Front Cell Neurosci 2021; 14:620379. [PMID: 33519385 PMCID: PMC7838331 DOI: 10.3389/fncel.2020.620379] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/08/2020] [Indexed: 01/19/2023] Open
Abstract
Neuronal migration is a fundamental brain development process that allows cells to move from their birthplaces to their sites of integration. Although neuronal migration largely ceases during embryonic and early postnatal development, neuroblasts continue to be produced and to migrate to a few regions of the adult brain such as the dentate gyrus and the subventricular zone (SVZ). In the SVZ, a large number of neuroblasts migrate into the olfactory bulb (OB) along the rostral migratory stream (RMS). Neuroblasts migrate in chains in a tightly organized micro-environment composed of astrocytes that ensheath the chains of neuroblasts and regulate their migration; the blood vessels that are used by neuroblasts as a physical scaffold and a source of molecular factors; and axons that modulate neuronal migration. In addition to diverse sets of extrinsic micro-environmental cues, long-distance neuronal migration involves a number of intrinsic mechanisms, including membrane and cytoskeleton remodeling, Ca2+ signaling, mitochondria dynamics, energy consumption, and autophagy. All these mechanisms are required to cope with the different micro-environment signals and maintain cellular homeostasis in order to sustain the proper dynamics of migrating neuroblasts and their faithful arrival in the target regions. Neuroblasts in the postnatal brain not only migrate into the OB but may also deviate from their normal path to migrate to a site of injury induced by a stroke or by certain neurodegenerative disorders. In this review, we will focus on the intrinsic mechanisms that regulate long-distance neuroblast migration in the adult brain and on how these pathways may be modulated to control the recruitment of neuroblasts to damaged/diseased brain areas.
Collapse
Affiliation(s)
- Cedric Bressan
- CERVO Brain Research Center, Quebec City, QC, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Quebec City, QC, Canada
| | - Armen Saghatelyan
- CERVO Brain Research Center, Quebec City, QC, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
63
|
Xia M, Zhao T, Wang X, Li Y, Li Y, Zheng T, Li J, Feng Y, Wei Y, Sun P. Brain-derived Neurotrophic Factor and Its Applications through Nanosystem Delivery. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 20:137-151. [PMID: 35194435 PMCID: PMC8842625 DOI: 10.22037/ijpr.2021.115705.15484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a protein that performs a neurotrophic function. BDNF and its receptors are widely expressed in the nervous system and can promote the growth of neurons and the formation of neuronal synapses in the brain. Studies have shown that a lack of BDNF can lead to impairment of memory and cognitive functions, indicating that BDNF plays an important role in mental illness and neurodegenerative diseases. The combination of stem cells and BDNF-releasing nanomaterials holds great promise in regenerative medicine, especially in the treatment of neurological diseases. For example, Alzheimer's disease, depression, Parkinson's disease, spinal cord injury, etc. The combination of stem cell/pharmacologically active carrier and BDNF-nano/hydrogel provided a useful new type of local delivery tool for the treatment of the nervous system and other diseases. It can not only provide BDNF but also stem cells. These studies will provide a scientific basis for the development and application of BDNF in the future.
Collapse
Affiliation(s)
- Mengyao Xia
- Department of Pharmacology, School of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji’nan 250355, China.
| | - Tingting Zhao
- Center for Foreign Language Translation, College of Foreign Languages, Shandong University of Traditional Chinese Medicine, Ji’nan250355, China.
| | - Xiaolong Wang
- Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji’nan 250355, China.
| | - Yang Li
- Department of Drug Design, College of Intelligence and Information Engineering, Shandong University of Traditional Chinese Medicine, Ji’nan 250355, China.
| | - Yanling Li
- Department of Pharmacology, School of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji’nan 250355, China.
| | - Tingting Zheng
- Department of Pharmacology, School of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji’nan 250355, China.
| | - Jiaxin Li
- Department of Pharmacology, School of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji’nan 250355, China.
| | - Yu Feng
- Department of Pharmacology, School of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji’nan 250355, China.
| | - Yongli Wei
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Ji’nan 250014, China.,Corresponding author:E-mail: ,
| | - Peng Sun
- Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji’nan 250355, China. ,Corresponding author:E-mail: ,
| |
Collapse
|
64
|
Alam JJ, Krakovsky M, Germann U, Levy A. Continuous administration of a p38α inhibitor during the subacute phase after transient ischemia-induced stroke in the rat promotes dose-dependent functional recovery accompanied by increase in brain BDNF protein level. PLoS One 2020; 15:e0233073. [PMID: 33275615 PMCID: PMC7717516 DOI: 10.1371/journal.pone.0233073] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022] Open
Abstract
There is unmet need for effective stroke therapies. Numerous neuroprotection attempts for acute cerebral ischemia have failed and as a result there is growing interest in developing therapies to promote functional recovery through increasing synaptic plasticity. For this research study, we hypothesized that in addition to its previously reported role in mediating cell death during the acute phase, the alpha isoform of p38 mitogen-activated protein kinase, p38α, may also contribute to interleukin-1β-mediated impairment of functional recovery during the subacute phase after acute ischemic stroke. Accordingly, an oral, brain-penetrant, small molecule p38α inhibitor, neflamapimod, was evaluated as a subacute phase stroke treatment to promote functional recovery. Neflamapimod administration to rats after transient middle cerebral artery occlusion at two dose levels was initiated outside of the previously characterized therapeutic window for neuroprotection of less than 24 hours for p38α inhibitors. Six-week administration of neflamapimod, starting at 48 hours after reperfusion, significantly improved behavioral outcomes assessed by the modified neurological severity score at Week 4 and at Week 6 post stroke in a dose-dependent manner. Neflamapimod demonstrated beneficial effects on additional measures of sensory and motor function. It also resulted in a dose-related increase in brain-derived neurotrophic factor (BDNF) protein levels, a previously reported potential marker of synaptic plasticity that was measured in brain homogenates at sacrifice. Taken together with literature evidence on the role of p38α-dependent suppression by interleukin-1β of BDNF-mediated synaptic plasticity and BDNF production, our findings support a mechanistic model in which inhibition of p38α promotes functional recovery after ischemic stroke by blocking the deleterious effects of interleukin-1β on synaptic plasticity. The dose-related in vivo efficacy of neflamapimod offers the possibility of having a therapy for stroke that could be initiated outside the short time window for neuroprotection and for improving recovery after a completed stroke.
Collapse
Affiliation(s)
- John J. Alam
- EIP Pharma, Inc., Boston, Massachusetts, United States of America
- * E-mail:
| | | | - Ursula Germann
- EIP Pharma, Inc., Boston, Massachusetts, United States of America
| | | |
Collapse
|
65
|
Huang F, Chen T, Chang J, Zhang C, Liao F, Wu L, Wang W, Yin Z. A conductive dual-network hydrogel composed of oxidized dextran and hyaluronic-hydrazide as BDNF delivery systems for potential spinal cord injury repair. Int J Biol Macromol 2020; 167:434-445. [PMID: 33278434 DOI: 10.1016/j.ijbiomac.2020.11.206] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 11/27/2020] [Accepted: 11/29/2020] [Indexed: 12/17/2022]
Abstract
Spinal cord injury (SCI) often causes neuronal death and axonal degeneration. In this study, we report a new strategy for preparing injectable and conductive polysaccharides-based hydrogels that could sustainably deliver brain-derived neurotrophic factor (BDNF) for SCI repair. We used poly(lactic-co-glycolic acid) (PLGA) as a carrier to encapsulate BDNF. The resulting microspheres were then modified with tannic acid (TA). The polysaccharides-based hydrogel composed of oxidized dextran (Dex) and hyaluronic acid-hydrazide (HA) was mixed with TA-modified microspheres to form the ultimate BDNF@TA-PLGA/Dex-HA hydrogel. Our results showed that the hydrogel had properties similar to natural spinal cords. Specifically, the hydrogel had soft mechanical properties and high electrical conductivity. The cross-sectional morphology of the hydrogel exhibited a continuous and porous structure. The swelling and degradation behaviors of the Dex-HA hydrogel in vitro indicated the incorporation of TA into hydrogel matrix could improve the stability of the hydrogel matrix as well as extend the release time of BDNF from the matrix. Furthermore, results from immunostaining and real-time PCR demonstrated that BDNF@TA-PLGA/Dex-HA hydrogel could promote the differentiation of neural stem cells (NSCs) into neurons and inhibit astrocyte differentiation in vitro. These results show the great potential of this hydrogel as a biomimetic material in SCI regeneration.
Collapse
Affiliation(s)
- Fei Huang
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Taiying Chen
- Department of Liver Transplantation, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Jun Chang
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Chi Zhang
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Faxue Liao
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Linwei Wu
- Department of Liver Transplantation, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China.
| | - Wenbin Wang
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Zongsheng Yin
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
66
|
Lacalle-Aurioles M, Cassel de Camps C, Zorca CE, Beitel LK, Durcan TM. Applying hiPSCs and Biomaterials Towards an Understanding and Treatment of Traumatic Brain Injury. Front Cell Neurosci 2020; 14:594304. [PMID: 33281561 PMCID: PMC7689345 DOI: 10.3389/fncel.2020.594304] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of disability and mortality in children and young adults and has a profound impact on the socio-economic wellbeing of patients and their families. Initially, brain damage is caused by mechanical stress-induced axonal injury and vascular dysfunction, which can include hemorrhage, blood-brain barrier disruption, and ischemia. Subsequent neuronal degeneration, chronic inflammation, demyelination, oxidative stress, and the spread of excitotoxicity can further aggravate disease pathology. Thus, TBI treatment requires prompt intervention to protect against neuronal and vascular degeneration. Rapid advances in the field of stem cells (SCs) have revolutionized the prospect of repairing brain function following TBI. However, more than that, SCs can contribute substantially to our knowledge of this multifaced pathology. Research, based on human induced pluripotent SCs (hiPSCs) can help decode the molecular pathways of degeneration and recovery of neuronal and glial function, which makes these cells valuable tools for drug screening. Additionally, experimental approaches that include hiPSC-derived engineered tissues (brain organoids and bio-printed constructs) and biomaterials represent a step forward for the field of regenerative medicine since they provide a more suitable microenvironment that enhances cell survival and grafting success. In this review, we highlight the important role of hiPSCs in better understanding the molecular pathways of TBI-related pathology and in developing novel therapeutic approaches, building on where we are at present. We summarize some of the most relevant findings for regenerative therapies using biomaterials and outline key challenges for TBI treatments that remain to be addressed.
Collapse
Affiliation(s)
- María Lacalle-Aurioles
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| | - Camille Cassel de Camps
- Department of Biological and Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Cornelia E Zorca
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| | - Lenore K Beitel
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| | - Thomas M Durcan
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| |
Collapse
|
67
|
Ge LJ, Yang FH, Li W, Wang T, Lin Y, Feng J, Chen NH, Jiang M, Wang JH, Hu XT, Chen G. In vivo Neuroregeneration to Treat Ischemic Stroke Through NeuroD1 AAV-Based Gene Therapy in Adult Non-human Primates. Front Cell Dev Biol 2020; 8:590008. [PMID: 33224952 PMCID: PMC7674285 DOI: 10.3389/fcell.2020.590008] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/09/2020] [Indexed: 12/15/2022] Open
Abstract
Stroke may cause severe death and disability but many clinical trials have failed in the past, partially because the lack of an effective method to regenerate new neurons after stroke. In this study, we report an in vivo neural regeneration approach through AAV NeuroD1-based gene therapy to repair damaged brains after ischemic stroke in adult non-human primates (NHPs). We demonstrate that ectopic expression of a neural transcription factor NeuroD1 in the reactive astrocytes after monkey cortical stroke can convert 90% of the infected astrocytes into neurons. Interestingly, astrocytes are not depleted in the NeuroD1-converted areas, consistent with the proliferative capability of astrocytes. Following ischemic stroke in monkey cortex, the NeuroD1-mediated astrocyte-to-neuron (AtN) conversion significantly increased local neuronal density, reduced microglia and macrophage, and surprisingly protected parvalbumin interneurons in the converted areas. Furthermore, the NeuroD1 gene therapy showed a broad time window in AtN conversion, from 10 to 30 days following ischemic stroke. The cortical astrocyte-converted neurons showed Tbr1+ cortical neuron identity, similar to our earlier findings in rodent animal models. Unexpectedly, NeuroD1 expression in converted neurons showed a significant decrease after 6 months of viral infection, indicating a downregulation of NeuroD1 after neuronal maturation in adult NHPs. These results suggest that in vivo cell conversion through NeuroD1-based gene therapy may be an effective approach to regenerate new neurons for tissue repair in adult primate brains.
Collapse
Affiliation(s)
- Long-Jiao Ge
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, China
| | - Fu-Han Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Wen Li
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Tao Wang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Yu Lin
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jie Feng
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Nan-Hui Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Min Jiang
- State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jian-Hong Wang
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Xin-Tian Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Shanghai, China
| | - Gong Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- Department of Biology, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
68
|
Neuroprotective Effects of Anti-proBDNF in a Rat Photothrombotic Ischemic Model. Neuroscience 2020; 446:261-270. [PMID: 32798590 DOI: 10.1016/j.neuroscience.2020.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 11/23/2022]
Abstract
Up-regulation of proBDNF in ischemic brain and the detrimental role of proBDNF on cellular survival has already been established. We propose that the up-regulated proBDNF may trigger the harmful events and evoke a secondary ischemic damage after ischemia. This study aimed to establish the neuroprotective effects of anti-proBDNF antibody in a rat photothrombotic ischemic model. Photothrombotic ischemic model was performed on Sprague Dawley rats and anti-proBDNF antibodies were administered intraperitoneally to the ischemic rats at a dose of 5 mg/kg after 6 hours (6 h) and on 3 days (3d) after ischemia. Behavioural tests were performed for sensorimotor functional analyses. Animals were euthanized at 7d for histochemical and biochemical studies. We observed higher proBDNF expression around the ischemic infarct. Higher level of apoptosis and inflammation was evident at 7d after ischemia on brain sections. Interestingly, the anti-proBDNF treatment instigated significant reduction of the infarction size as detected by Haematoxylin and Eosin (H&E) staining. Similar reduction of apoptotic signaling proteins in western blot and immunostaining after anti-proBDNF treatment was found. Up-regulation of synaptic protein expression was also observed after this treatment. Significant sensorimotor functional improvements were also noticed at 7d after anti-proBDNF treatment. We conclude that anti-proBDNF treatment is anti-apoptotic and anti-inflammatory, and plays advantageous role in promoting cellular growth and improving sensorimotor function after ischemic insult. Taken together, our study suggests that this anti-proBDNF treatment can be considered as a therapeutic approach for ischemic recovery.
Collapse
|
69
|
Wilson KL, Carmichael ST, Segura T. Injection of Hydrogel Biomaterial Scaffolds to The Brain After Stroke. J Vis Exp 2020:10.3791/61450. [PMID: 33074256 PMCID: PMC8516153 DOI: 10.3791/61450] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Stroke is the leading cause of disability and the fifth-leading cause of death in the United States. Approximately 87% of all strokes are ischemic strokes and are defined as the sudden blockage of a vessel supplying blood to the brain. Within minutes of the blockage, cells begin to die and result in irreparable tissue damage. Current therapeutic treatments focus on clot removal or lysis to allow for the reperfusion and prevent more severe brain damage. Although transient brain plasticity may salvage some of the damaged tissue over time, significant fractions of patients are left with neurological deficits that will never resolve. There is a lack of therapeutic options to treat neurological deficits caused by stroke, emphasizing the need to develop new strategies to treat this growing patient population. Injectable biomaterials are currently being designed to enhance brain plasticity and improve endogenous repair through the delivery of active agents or stem cells. One method to test these approaches is to utilize a rodent stroke model, inject the biomaterial into the stroke core, and assess repair. Knowing the precise location of the stroke core is imperative for the accurate treatment after stroke, therefore, a stroke model that results in a predictable stroke location is preferable to avoid the need for imaging prior to injection. The following protocol will cover how to induce a photothrombotic stroke, how to inject a hydrogel in a controlled and precise manner, and how to extract and cryosection the brain while keeping the biomaterial intact. In addition, we will highlight how these same hydrogel materials can be used for the co-delivery of stem cells. This protocol can be generalized to the use of other injectable biomaterials into the stroke core.
Collapse
Affiliation(s)
| | | | - Tatiana Segura
- Department of Biomedical Engineering, Duke University; Department of Neurology, Duke University; Department of Dermatology, Duke University;
| |
Collapse
|
70
|
Sequeira E, Pierce ML, Akasheh D, Sellers S, Gerwick WH, Baden DG, Murray TF. Epicortical Brevetoxin Treatment Promotes Neural Repair and Functional Recovery after Ischemic Stroke. Mar Drugs 2020; 18:md18070374. [PMID: 32708077 PMCID: PMC7404386 DOI: 10.3390/md18070374] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/17/2020] [Accepted: 07/17/2020] [Indexed: 02/01/2023] Open
Abstract
Emerging literature suggests that after a stroke, the peri-infarct region exhibits dynamic changes in excitability. In rodent stroke models, treatments that enhance excitability in the peri-infarct cerebral cortex promote motor recovery. This increase in cortical excitability and plasticity is opposed by increases in tonic GABAergic inhibition in the peri-infarct zone beginning three days after a stroke in a mouse model. Maintenance of a favorable excitatory-inhibitory balance promoting cerebrocortical excitability could potentially improve recovery. Brevetoxin-2 (PbTx-2) is a voltage-gated sodium channel (VGSC) gating modifier that increases intracellular sodium ([Na+]i), upregulates N-methyl-D-aspartate receptor (NMDAR) channel activity and engages downstream calcium (Ca2+) signaling pathways. In immature cerebrocortical neurons, PbTx-2 promoted neuronal structural plasticity by increasing neurite outgrowth, dendritogenesis and synaptogenesis. We hypothesized that PbTx-2 may promote excitability and structural remodeling in the peri-infarct region, leading to improved functional outcomes following a stroke. We tested this hypothesis using epicortical application of PbTx-2 after a photothrombotic stroke in mice. We show that PbTx-2 enhanced the dendritic arborization and synapse density of cortical layer V pyramidal neurons in the peri-infarct cortex. PbTx-2 also produced a robust improvement of motor recovery. These results suggest a novel pharmacologic approach to mimic activity-dependent recovery from stroke.
Collapse
Affiliation(s)
- Erica Sequeira
- Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE 68123, USA; (E.S.); (M.L.P.); (D.A.); (S.S.)
| | - Marsha L. Pierce
- Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE 68123, USA; (E.S.); (M.L.P.); (D.A.); (S.S.)
| | - Dina Akasheh
- Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE 68123, USA; (E.S.); (M.L.P.); (D.A.); (S.S.)
| | - Stacey Sellers
- Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE 68123, USA; (E.S.); (M.L.P.); (D.A.); (S.S.)
| | - William H. Gerwick
- Center for Marine Biotechnology & Biomedicine, Scripps Institution of Oceanography, San Diego, La Jolla, CA 92093, USA;
| | - Daniel G. Baden
- Center for Marine Science University of North Carolina Wilmington, Wilmington, NC 28409, USA;
| | - Thomas F. Murray
- Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE 68123, USA; (E.S.); (M.L.P.); (D.A.); (S.S.)
- Correspondence:
| |
Collapse
|
71
|
Rey F, Barzaghini B, Nardini A, Bordoni M, Zuccotti GV, Cereda C, Raimondi MT, Carelli S. Advances in Tissue Engineering and Innovative Fabrication Techniques for 3-D-Structures: Translational Applications in Neurodegenerative Diseases. Cells 2020; 9:cells9071636. [PMID: 32646008 PMCID: PMC7407518 DOI: 10.3390/cells9071636] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/01/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022] Open
Abstract
In the field of regenerative medicine applied to neurodegenerative diseases, one of the most important challenges is the obtainment of innovative scaffolds aimed at improving the development of new frontiers in stem-cell therapy. In recent years, additive manufacturing techniques have gained more and more relevance proving the great potential of the fabrication of precision 3-D scaffolds. In this review, recent advances in additive manufacturing techniques are presented and discussed, with an overview on stimulus-triggered approaches, such as 3-D Printing and laser-based techniques, and deposition-based approaches. Innovative 3-D bioprinting techniques, which allow the production of cell/molecule-laden scaffolds, are becoming a promising frontier in disease modelling and therapy. In this context, the specific biomaterial, stiffness, precise geometrical patterns, and structural properties are to be considered of great relevance for their subsequent translational applications. Moreover, this work reports numerous recent advances in neural diseases modelling and specifically focuses on pre-clinical and clinical translation for scaffolding technology in multiple neurodegenerative diseases.
Collapse
Affiliation(s)
- Federica Rey
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Via Grassi 74, 20157 Milan, Italy; (F.R.); (G.V.Z.)
- Pediatric Clinical Research Center Fondazione “Romeo ed Enrica Invernizzi”, University of Milano, Via Grassi 74, 20157 Milano, Italy
| | - Bianca Barzaghini
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy; (B.B.); (A.N.)
| | - Alessandra Nardini
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy; (B.B.); (A.N.)
| | - Matteo Bordoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy;
| | - Gian Vincenzo Zuccotti
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Via Grassi 74, 20157 Milan, Italy; (F.R.); (G.V.Z.)
- Pediatric Clinical Research Center Fondazione “Romeo ed Enrica Invernizzi”, University of Milano, Via Grassi 74, 20157 Milano, Italy
| | - Cristina Cereda
- Genomic and post-Genomic Center, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy;
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy; (B.B.); (A.N.)
- Correspondence: (M.T.R.); (S.C.); Tel.: +390-223-994-306 (M.T.R.); +390-250-319-825 (S.C.)
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Via Grassi 74, 20157 Milan, Italy; (F.R.); (G.V.Z.)
- Pediatric Clinical Research Center Fondazione “Romeo ed Enrica Invernizzi”, University of Milano, Via Grassi 74, 20157 Milano, Italy
- Correspondence: (M.T.R.); (S.C.); Tel.: +390-223-994-306 (M.T.R.); +390-250-319-825 (S.C.)
| |
Collapse
|
72
|
He W, Reaume M, Hennenfent M, Lee BP, Rajachar R. Biomimetic hydrogels with spatial- and temporal-controlled chemical cues for tissue engineering. Biomater Sci 2020; 8:3248-3269. [PMID: 32490441 PMCID: PMC7323904 DOI: 10.1039/d0bm00263a] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Biomimetic hydrogels have emerged as the most useful tissue engineering scaffold materials. Their versatile chemistry can recapitulate multiple physical and chemical features to integrate cells, scaffolds, and signaling molecules for tissue regeneration. Due to their highly hydrophilic nature hydrogels can recreate nutrient-rich aqueous environments for cells. Soluble regulatory molecules can be incorporated to guide cell proliferation and differentiation. Importantly, the controlled dynamic parameters and spatial distribution of chemical cues in hydrogel scaffolds are critical for cell-cell communication, cell-scaffold interaction, and morphogenesis. Herein, we review biomimetic hydrogels that provide cells with spatiotemporally controlled chemical cues as tissue engineering scaffolds. Specifically, hydrogels with temporally controlled growth factor-release abilities, spatially controlled conjugated bioactive molecules/motifs, and targeting delivery and reload properties for tissue engineering applications are discussed in detail. Examples of hydrogels that possess clinically favorable properties, such as injectability, self-healing ability, stimulus-responsiveness, and pro-remodeling features, are also covered.
Collapse
Affiliation(s)
- Weilue He
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
- FM Wound Care, LLC, Hancock, MI 49930, USA
| | - Max Reaume
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Maureen Hennenfent
- Department of Civil and Environmental Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Bruce P. Lee
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Rupak Rajachar
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| |
Collapse
|
73
|
Abstract
Stroke is the leading cause of long-term disability with no current treatment addressing post-stroke disability. The complex pathophysiology of stroke and the brain's limited potential for regeneration prevents sufficient endogenous repair for complete recovery. While engineered materials provide an exciting opportunity to augment endogenous repair in conjunction with other therapies that address post-stroke disability, much of the preclinical work in this arena is still in its infancy. Biomaterials can be used to enhance drug- or stem cell-sustained and targeted delivery. Moreover, materials can act as extracellular matrix-mimics and augment a pro-repair environment by addressing astrogliosis, inflammation, neurogenesis, axonal sprouting, and angiogenesis. Lastly, there is a growing need to elucidate stroke repair mechanisms to identify novel targets to inform material design for brain repair after stroke.
Collapse
Affiliation(s)
- Kevin Erning
- Duke University Biomedical Engineering Department, 101 Science Drive, CIEMAS, NC 27707
| | - Tatiana Segura
- Duke University Biomedical Engineering Department, 101 Science Drive, CIEMAS, NC 27707
| |
Collapse
|
74
|
Phosphodiesterase 10A Inhibition Leads to Brain Region-Specific Recovery Based on Stroke Type. Transl Stroke Res 2020; 12:303-315. [PMID: 32378029 PMCID: PMC7644574 DOI: 10.1007/s12975-020-00819-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/19/2020] [Accepted: 04/22/2020] [Indexed: 01/08/2023]
Abstract
Stroke is the leading cause of adult disability. Recovery of function after stroke involves signaling events that are mediated by cAMP and cGMP pathways, such as axonal sprouting, neurogenesis, and synaptic plasticity. cAMP and cGMP are degraded by phosphodiesterases (PDEs), which are differentially expressed in brain regions. PDE10A is highly expressed in the basal ganglia/striatum. We tested a novel PDE10A inhibitor (TAK-063) for its effects on functional recovery. Stroke was produced in mice in the cortex or the striatum. Behavioral recovery was measured to 9 weeks. Tissue outcome measures included analysis of growth factor levels, angiogenesis, neurogenesis, gliogenesis, and inflammation. TAK-063 improved motor recovery after striatal stroke in a dose-related manner, but not in cortical stroke. Recovery of motor function correlated with increases in striatal brain-derived neurotrophic factor. TAK-063 treatment also increased motor system axonal connections. Stroke affects distinct brain regions, with each comprising different cellular and molecular elements. Inhibition of PDE10A improved recovery of function after striatal but not cortical stroke, consistent with its brain localization. This experiment is the first demonstration of brain region-specific enhanced functional recovery after stroke, and indicates that differential molecular signaling between brain regions can be exploited to improve recovery based on stroke subtype.
Collapse
|
75
|
Design and evaluation of a biosynthesized cellulose drug releasing duraplasty. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110677. [DOI: 10.1016/j.msec.2020.110677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 12/20/2019] [Accepted: 01/18/2020] [Indexed: 12/16/2022]
|
76
|
González-Nieto D, Fernández-Serra R, Pérez-Rigueiro J, Panetsos F, Martinez-Murillo R, Guinea GV. Biomaterials to Neuroprotect the Stroke Brain: A Large Opportunity for Narrow Time Windows. Cells 2020; 9:E1074. [PMID: 32357544 PMCID: PMC7291200 DOI: 10.3390/cells9051074] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke represents one of the most prevalent pathologies in humans and is a leading cause of death and disability. Anti-thrombolytic therapy with tissue plasminogen activator (t-PA) and surgical thrombectomy are the primary treatments to recanalize occluded vessels and normalize the blood flow in ischemic and peri-ischemic regions. A large majority of stroke patients are refractory to treatment or are not eligible due to the narrow time window of therapeutic efficacy. In recent decades, we have significantly increased our knowledge of the molecular and cellular mechanisms that inexorably lead to progressive damage in infarcted and peri-lesional brain areas. As a result, promising neuroprotective targets have been identified and exploited in several stroke models. However, these considerable advances have been unsuccessful in clinical contexts. This lack of clinical translatability and the emerging use of biomaterials in different biomedical disciplines have contributed to developing a new class of biomaterial-based systems for the better control of drug delivery in cerebral disorders. These systems are based on specific polymer formulations structured in nanoparticles and hydrogels that can be administered through different routes and, in general, bring the concentrations of drugs to therapeutic levels for prolonged times. In this review, we first provide the general context of the molecular and cellular mechanisms impaired by cerebral ischemia, highlighting the role of excitotoxicity, inflammation, oxidative stress, and depolarization waves as the main pathways and targets to promote neuroprotection avoiding neuronal dysfunction. In the second part, we discuss the versatile role played by distinct biomaterials and formats to support the sustained administration of particular compounds to neuroprotect the cerebral tissue at risk of damage.
Collapse
Affiliation(s)
- Daniel González-Nieto
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (R.F.-S.); (J.P.-R.); (G.V.G.)
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Rocío Fernández-Serra
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (R.F.-S.); (J.P.-R.); (G.V.G.)
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - José Pérez-Rigueiro
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (R.F.-S.); (J.P.-R.); (G.V.G.)
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Fivos Panetsos
- Neurocomputing and Neurorobotics Research Group: Faculty of Biology and Faculty of Optics, Universidad Complutense de Madrid, 28040 Madrid, Spain;
- Brain Plasticity Group, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | | | - Gustavo V. Guinea
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (R.F.-S.); (J.P.-R.); (G.V.G.)
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| |
Collapse
|
77
|
Injectable hydrogel enables local and sustained co-delivery to the brain: Two clinically approved biomolecules, cyclosporine and erythropoietin, accelerate functional recovery in rat model of stroke. Biomaterials 2020; 235:119794. [DOI: 10.1016/j.biomaterials.2020.119794] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 01/06/2020] [Accepted: 01/15/2020] [Indexed: 12/20/2022]
|
78
|
Tsintou M, Dalamagkas K, Makris N. Taking central nervous system regenerative therapies to the clinic: curing rodents versus nonhuman primates versus humans. Neural Regen Res 2020; 15:425-437. [PMID: 31571651 PMCID: PMC6921352 DOI: 10.4103/1673-5374.266048] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/04/2019] [Indexed: 12/17/2022] Open
Abstract
The central nervous system is known to have limited regenerative capacity. Not only does this halt the human body's reparative processes after central nervous system lesions, but it also impedes the establishment of effective and safe therapeutic options for such patients. Despite the high prevalence of stroke and spinal cord injury in the general population, these conditions remain incurable and place a heavy burden on patients' families and on society more broadly. Neuroregeneration and neural engineering are diverse biomedical fields that attempt reparative treatments, utilizing stem cells-based strategies, biologically active molecules, nanotechnology, exosomes and highly tunable biodegradable systems (e.g., certain hydrogels). Although there are studies demonstrating promising preclinical results, safe clinical translation has not yet been accomplished. A key gap in clinical translation is the absence of an ideal animal or ex vivo model that can perfectly simulate the human microenvironment, and also correspond to all the complex pathophysiological and neuroanatomical factors that affect functional outcomes in humans after central nervous system injury. Such an ideal model does not currently exist, but it seems that the nonhuman primate model is uniquely qualified for this role, given its close resemblance to humans. This review considers some regenerative therapies for central nervous system repair that hold promise for future clinical translation. In addition, it attempts to uncover some of the main reasons why clinical translation might fail without the implementation of nonhuman primate models in the research pipeline.
Collapse
Affiliation(s)
- Magdalini Tsintou
- Departments of Psychiatry and Neurology Services, Center for Neural Systems Investigations, Center for Morphometric Analysis, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- University College of London Division of Surgery & Interventional Science, Center for Nanotechnology & Regenerative Medicine, University College London, London, UK
| | - Kyriakos Dalamagkas
- University College of London Division of Surgery & Interventional Science, Center for Nanotechnology & Regenerative Medicine, University College London, London, UK
- Department of Physical Medicine and Rehabilitation, The University of Texas Health Science Center at Houston, Houston, TX, USA
- The Institute for Rehabilitation and Research Memorial Hermann Research Center, The Institute for Rehabilitation and Research Memorial Hermann Hospital, Houston, TX, USA
| | - Nikos Makris
- Departments of Psychiatry and Neurology Services, Center for Neural Systems Investigations, Center for Morphometric Analysis, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
79
|
McCrary MR, Jesson K, Wei ZZ, Logun M, Lenear C, Tan S, Gu X, Jiang MQ, Karumbaiah L, Ping Yu S, Wei L. Cortical Transplantation of Brain-Mimetic Glycosaminoglycan Scaffolds and Neural Progenitor Cells Promotes Vascular Regeneration and Functional Recovery after Ischemic Stroke in Mice. Adv Healthc Mater 2020; 9:e1900285. [PMID: 31977165 PMCID: PMC7358896 DOI: 10.1002/adhm.201900285] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 01/08/2020] [Indexed: 12/14/2022]
Abstract
Stroke causes significant mortality and morbidity. Currently, there are no treatments which can regenerate brain tissue lost to infarction. Neural progenitor cells (NPCs) are at the forefront of preclinical studies for regenerative stroke therapies. NPCs can differentiate into and replace neurons and promote endogenous recovery mechanisms such as angiogenesis via trophic factor production and release. The stroke core is hypothetically the ideal location for replacement of neural tissue since it is in situ and develops into a potential space where injections may be targeted with minimal compression of healthy peri-infarct tissue. However, the compromised perfusion and tissue degradation following ischemia create an inhospitable environment resistant to cellular therapy. Overcoming these limitations is critical to advancing cellular therapy. In this work, the therapeutic potential of mouse-induced pluripotent stem cell derived NPCs is tested encapsulated in a basic fibroblast growth factor (bFGF) binding chondroitin sulfate-A (CS-A) hydrogel transplanted into the infarct core in a mouse sensorimotor cortex mini-stroke model. It is shown that CS-A encapsulation significantly improves vascular remodeling, cortical blood flow, and sensorimotor behavioral outcomes after stroke. It is found these improvements are negated by blocking bFGF, suggesting that the sustained trophic signaling endowed by the CS-A hydrogel combined with NPC transplantation can promote tissue repair.
Collapse
Affiliation(s)
- Myles R. McCrary
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Kaleena Jesson
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Zheng Z. Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Meghan Logun
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
| | - Christopher Lenear
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
| | - Stephen Tan
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Michael Q. Jiang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA 30033, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|
80
|
Brain-Derived Neurotrophic Factor and Its Potential Therapeutic Role in Stroke Comorbidities. Neural Plast 2020; 2020:1969482. [PMID: 32399020 PMCID: PMC7204205 DOI: 10.1155/2020/1969482] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/14/2019] [Accepted: 11/08/2019] [Indexed: 12/17/2022] Open
Abstract
With the rise in the aging global population, stroke comorbidities have become a serious health threat and a tremendous economic burden on human society. Current therapeutic strategies mainly focus on protecting neurons from cytotoxic damage at the acute phase upon stroke onset, which not only is a difficult way to ameliorate stroke symptoms but also presents a challenge for the patients to receive effective treatment in time. The brain-derived neurotrophic factor (BDNF) is the most abundant neurotrophin in the adult brain, which possesses a remarkable capability to repair brain damage. Recent promising preclinical outcomes have made BDNF a popular late-stage target in the development of novel stroke treatments. In this review, we aim to summarize the latest progress in the understanding of the cellular/molecular mechanisms underlying stroke pathogenesis, current strategies and difficulties in drug development, the mechanism of BDNF action in poststroke neurorehabilitation and neuroplasticity, and recent updates in novel therapeutic methods.
Collapse
|
81
|
Nozohouri S, Sifat AE, Vaidya B, Abbruscato TJ. Novel approaches for the delivery of therapeutics in ischemic stroke. Drug Discov Today 2020; 25:535-551. [PMID: 31978522 DOI: 10.1016/j.drudis.2020.01.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/20/2019] [Accepted: 01/15/2020] [Indexed: 02/06/2023]
Abstract
Here, we review novel approaches to deliver neuroprotective drugs to salvageable penumbral brain areas of stroke injury with the goals of offsetting ischemic brain injury and enhancing recovery.
Collapse
Affiliation(s)
- Saeideh Nozohouri
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Ali Ehsan Sifat
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Bhuvaneshwar Vaidya
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| |
Collapse
|
82
|
The use of bioactive matrices in regenerative therapies for traumatic brain injury. Acta Biomater 2020; 102:1-12. [PMID: 31751809 DOI: 10.1016/j.actbio.2019.11.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/22/2019] [Accepted: 11/13/2019] [Indexed: 01/01/2023]
Abstract
Functional deficits due to neuronal loss are a common theme across multiple neuropathologies, including traumatic brain injury (TBI). Apart from mitigating cell death, another approach to treating brain injuries involves re-establishing the neural circuitry at the lesion site by utilizing exogeneous and/or endogenous stem cells to achieve functional recovery. While there has been limited success, the emergence of new bioactive matrices that promote neural repair introduces new perspectives on the development of regenerative therapies for TBI. This review briefly discusses current development on cell-based therapies and the use of bioactive matrices, hydrogels in particular, when incorporated in regenerative therapies. Desirable characteristics of bioactive matrices that have been shown to augment neural repair in TBI models were identified and further discussed. Understanding the relative outcomes of newly developed biomaterials implanted in vivo can better guide the development of biomaterials as a therapeutic strategy, for biomaterial-based cellular therapies are still in their nascent stages. Nonetheless, the value of bioactive matrices as a treatment for acute brain injuries should be appreciated and further developed. STATEMENT OF SIGNIFICANCE: Cell-based therapies have received attention as an alternative therapeutic strategy to improve clinical outcome post-traumatic brain injury but have achieved limited success. Whilst the incorporation of newly developed biomaterials in regenerative therapies has shown promise in augmenting neural repair, studies have revealed new hurdles which must be overcome to improve their therapeutic efficacy. This review discusses the recent development of cell-based therapies with a specific focus on the use of bioactive matrices in the form of hydrogels, to complement cell transplantation within the injured brain. Moreover, this review consolidates in vivo animal studies that demonstrate relative functional outcome upon the implantation of different biomaterials to highlight their desirable traits to guide their development for regenerative therapies in traumatic brain injury.
Collapse
|
83
|
Motor deficit in the mouse ferric chloride-induced distal middle cerebral artery occlusion model of stroke. Behav Brain Res 2019; 380:112418. [PMID: 31812504 DOI: 10.1016/j.bbr.2019.112418] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/22/2022]
Abstract
Ferric chloride-induced distal middle cerebral artery occlusion (MCAO) model of stroke was described in mice several years ago, however it lacked in-depth evaluation of the post-stroke functional outcomes in the animals. In this study, we reproduced the recently developed model and expanded its characterization by thorough evaluation of blood supply, cerebral infarction, and motor function in adult male and female mice up to 14 days after stroke. Our observations indicate near complete interruption of blood flow in the distal MCA shortly after application of 20 % ferric chloride over the artery through a cranial window, which remained occluded for at least 4 h. As expected, infarction of the brain tissue, documented by TTC and hematoxylin stains, was restricted to the cerebral cortex. We also systematically evaluated motor impairment of the animals in this model. For this, a series of studies were carried out in male and female mice up to 14 days after stroke, and motor function was assessed in cylinder and grid-walking tests in blinded manner. Contrary to our expectations, the results of both motor tests indicated minor, transient motor deficit in mice after stroke. Based on these observations, we conclude that the mouse ferric chloride-induced distal MCAO model is likely not suitable for proof-of-concept and preclinical studies where motor function is an important outcome measure.
Collapse
|
84
|
Belousov A, Titov S, Shved N, Garbuz M, Malykin G, Gulaia V, Kagansky A, Kumeiko V. The Extracellular Matrix and Biocompatible Materials in Glioblastoma Treatment. Front Bioeng Biotechnol 2019; 7:341. [PMID: 31803736 PMCID: PMC6877546 DOI: 10.3389/fbioe.2019.00341] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 10/30/2019] [Indexed: 02/06/2023] Open
Abstract
During cancer genesis, the extracellular matrix (ECM) in the human brain undergoes important transformations, starting to resemble embryonic brain cell milieu with a much denser structure. However, the stiffness of the tumor ECM does not preclude cancer cells from migration. The importance of the ECM role in normal brain tissue as well as in tumor homeostasis has engaged much effort in trials to implement ECM as a target and an instrument in the treatment of brain cancers. This review provides a detailed analysis of both experimental and applied approaches in combined therapy for gliomas in adults. In general, matrix materials for glioma treatment should have properties facilitating the simplest delivery into the body. Hence, to deliver an artificial implant directly into the operation cavity it should be packed into a gel form, while for bloodstream injections matrix needs to be in the form of polymer micelles, nanoparticles, etc. Furthermore, the delivered material should mimic biomechanical properties of the native tissue, support vital functions, and slow down or stop the proliferation of surrounding cells for a prolonged period. The authors propose a two-step approach aimed, on the one hand, at elimination of remaining cancer cells and on the other hand, at restoring normal brain tissue. Thereby, the first bioartificial matrix to be applied should have relatively low elastic modulus should be loaded with anticancer drugs, while the second material with a higher elastic modulus for neurite outgrowth support should contain specific factors stimulating neuroregeneration.
Collapse
Affiliation(s)
- Andrei Belousov
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Sergei Titov
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- School of Natural Sciences, Far Eastern Federal University, Vladivostok, Russia
| | - Nikita Shved
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok, Russia
| | - Mikhail Garbuz
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Grigorii Malykin
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok, Russia
| | - Valeriia Gulaia
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Alexander Kagansky
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Vadim Kumeiko
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- School of Natural Sciences, Far Eastern Federal University, Vladivostok, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok, Russia
| |
Collapse
|
85
|
Wang J, Huang Q, Ding J, Wang X. Elevated serum levels of brain-derived neurotrophic factor and miR-124 in acute ischemic stroke patients and the molecular mechanism. 3 Biotech 2019; 9:386. [PMID: 31656724 PMCID: PMC6778548 DOI: 10.1007/s13205-019-1914-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 09/21/2019] [Indexed: 10/25/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and microRNAs (miRNAs) play a significant role in the pathogenesis of acute ischemic stroke (AIS). The present study investigates the elevated expression of BDNF and miR-124 in AIS patients. In the present study, serum samples from AIS patients and healthy controls were collected to determine the regulatory role and mechanism of operation of BDNF and to determine the regulatory miRNAs involved in AIS. Using bioinformatics analysis, we identified putative and regulatory miR-124. The effect of miR-124 on BDNF expression was examined in human neuronal cell lines. Moreover, the function of miR-124 in regulating BDNF was analyzed by assessing the serum level of BDNF in both AIS patients and healthy controls. The results indicate that the BDNF level of AIS patients is very low compared with that of controls. In contrast, real-time polymerase chain reaction (RT-PCR) data revealed a very high serum level of miR-124 in AIS patients relative to healthy individuals. The associations of the National Institutes of Health (NIH) stroke scale (NIHSS) score with BDNF and BDNF-related miR-124 serum levels were calculated using Pearson's/Spearman's correlation coefficient. The findings revealed a negative correlation between NIHSS score and BDNF level, whereas a positive correlation was observed between NIHSS score and miR-124. In addition, the relationship between serum BDNF and miR-124 was negative in AIS patients. In conclusion, this study provides strong evidence that serum BDNF and the BDNF-regulatory miR-124 may serve as molecular markers for AIS.
Collapse
Affiliation(s)
- Jie Wang
- Department of Neurology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, No 1111 of XianXia Road, Shanghai, 200335 China
| | - Qiong Huang
- Department of Neurology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, No 1111 of XianXia Road, Shanghai, 200335 China
| | - Ji Ding
- Department of Neurology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, No 1111 of XianXia Road, Shanghai, 200335 China
| | - Xiaoping Wang
- Department of Neurology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, No 1111 of XianXia Road, Shanghai, 200335 China
| |
Collapse
|
86
|
Vitronectin mitigates stroke-increased neurogenesis only in female mice and through FAK-regulated IL-6. Exp Neurol 2019; 323:113088. [PMID: 31678139 DOI: 10.1016/j.expneurol.2019.113088] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/03/2019] [Accepted: 10/20/2019] [Indexed: 12/18/2022]
Abstract
Vitronectin (VTN) is a blood protein produced mainly by the liver. We show that VTN leaks from the bloodstream into the injury site and neighboring subventricular zone (SVZ) following ischemic stroke (middle cerebral artery occlusion, MCAO) in adult mice. MCAO is known to increase neurogenesis after stroke. VTN inhibits this response in females, but not in males, as shown by ~70% more stroke-induced SVZ neurogenesis in female VTN-/- mice at 14 d. In female VTN-/- mice, stroke-induced expression of interleukin-6 (IL-6) at 24 h was reduced in the SVZ. The closely related leukemia inhibitory factor (LIF) or pro-neurogenic ciliary neurotrophic factor (CNTF) were not affected. The female-specific effect of VTN on IL-6 expression was not due to sex hormones, as shown by ovariectomy and castration. IL-6 injection next to the SVZ reversed the MCAO-induced increase in neurogenesis seen in VTN-/- mice. Our in vitro and vivo data suggest that plasma VTN activates focal adhesion kinase (FAK) in the SVZ following MCAO, which reduces IL-6 expression in astrocytes but increases it in other cells such as microglia/macrophages. Inducible conditional astrocytic FAK deletion increased MCAO-induced IL-6 expression in females at 24 h and blocked MCAO-induced neurogenesis at 14 d, confirming a key detrimental role of IL-6. Collectively, these data suggest that leakage of VTN into the SVZ reduces the neurogenic response to stroke in female mice by promoting IL-6 expression. Reducing VTN or VTN signaling may be an approach to promote neurogenesis for neuroprotection and cell replacement after stroke in females.
Collapse
|
87
|
Boltze J, Modo MM, Mays RW, Taguchi A, Jolkkonen J, Savitz SI. Stem Cells as an Emerging Paradigm in Stroke 4: Advancing and Accelerating Preclinical Research. Stroke 2019; 50:3299-3306. [PMID: 31658004 DOI: 10.1161/strokeaha.119.025436] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Johannes Boltze
- From the School of Life Sciences, University of Warwick, Coventry, United Kingdom (J.B.)
| | - Michel M Modo
- Departments of Radiology (M.M.M.), McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA.,Bioengineering (M.M.M.), McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA
| | - Robert W Mays
- Department of Neurosciences, Athersys, Inc, Cleveland, OH (R.W.M.)
| | - Akihiko Taguchi
- Department of Regenerative Medicine, Institute for Biomedical Research and Innovation, Kobe, Japan (A.T.)
| | - Jukka Jolkkonen
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland (J.J.).,Neurocenter, Kuopio University Hospital, Finland (J.J.).,A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (J.J.)
| | - Sean I Savitz
- Institute for Stroke and Cerebrovascular Disease, UTHealth, Houston, TX (S.I.S.)
| | | |
Collapse
|
88
|
Chan SJ, Niu W, Hayakawa K, Hamanaka G, Wang X, Cheah PS, Guo S, Yu Z, Arai K, Selim MH, Kurisawa M, Spector M, Lo EH. Promoting Neuro-Supportive Properties of Astrocytes with Epidermal Growth Factor Hydrogels. Stem Cells Transl Med 2019; 8:1242-1248. [PMID: 31483567 PMCID: PMC6877762 DOI: 10.1002/sctm.19-0159] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 07/30/2019] [Indexed: 12/24/2022] Open
Abstract
Biomaterials provide novel platforms to deliver stem cell and growth factor therapies for central nervous system (CNS) repair. The majority of these approaches have focused on the promotion of neural progenitor cells and neurogenesis. However, it is now increasingly recognized that glial responses are critical for recovery in the entire neurovascular unit. In this study, we investigated the cellular effects of epidermal growth factor (EGF) containing hydrogels on primary astrocyte cultures. Both EGF alone and EGF-hydrogel equally promoted astrocyte proliferation, but EGF-hydrogels further enhanced astrocyte activation, as evidenced by a significantly elevated Glial fibrillary acidic protein (GFAP) gene expression. Thereafter, conditioned media from astrocytes activated by EGF-hydrogel protected neurons against injury and promoted synaptic plasticity after oxygen-glucose deprivation. Taken together, these findings suggest that EGF-hydrogels can shift astrocytes into neuro-supportive phenotypes. Consistent with this idea, quantitative-polymerase chain reaction (qPCR) demonstrated that EGF-hydrogels shifted astrocytes in part by downregulating potentially negative A1-like genes (Fbln5 and Rt1-S3) and upregulating potentially beneficial A2-like genes (Clcf1, Tgm1, and Ptgs2). Further studies are warranted to explore the idea of using biomaterials to modify astrocyte behavior and thus indirectly augment neuroprotection and neuroplasticity in the context of stem cell and growth factor therapies for the CNS. Stem Cells Translational Medicine 2019;8:1242&1248.
Collapse
Affiliation(s)
- Su Jing Chan
- Neuroprotection Research Laboratory, Departments of Radiology and NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Wanting Niu
- Tissue Engineering LaboratoriesVA Boston Healthcare SystemBostonMassachusettsUSA,Department of Orthopaedic SurgeryBrigham & Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Kazuhide Hayakawa
- Neuroprotection Research Laboratory, Departments of Radiology and NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Gen Hamanaka
- Neuroprotection Research Laboratory, Departments of Radiology and NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Xiaoying Wang
- Neuroprotection Research Laboratory, Departments of Radiology and NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Pike See Cheah
- Neuroprotection Research Laboratory, Departments of Radiology and NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA,Genetics and Regenerative Medicine Research Center, Department of Human AnatomyUniversiti PutraSerdangSelangorMalaysia
| | - Shuzhen Guo
- Neuroprotection Research Laboratory, Departments of Radiology and NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Zhangyang Yu
- Neuroprotection Research Laboratory, Departments of Radiology and NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Magdy H. Selim
- Department of NeurologyBeth Israel Deaconess Medical Center, Harvard Medical SchoolBostonMassachusettsUSA
| | - Motoichi Kurisawa
- Institute Bioengineering and Nanotechnology, A*STARSingaporeSingapore
| | - Myron Spector
- Tissue Engineering LaboratoriesVA Boston Healthcare SystemBostonMassachusettsUSA,Department of Orthopaedic SurgeryBrigham & Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA,Harvard‐MIT Division of Health Sciences and TechnologyMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Eng H. Lo
- Neuroprotection Research Laboratory, Departments of Radiology and NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| |
Collapse
|
89
|
Bolan F, Louca I, Heal C, Cunningham CJ. The Potential of Biomaterial-Based Approaches as Therapies for Ischemic Stroke: A Systematic Review and Meta-Analysis of Pre-clinical Studies. Front Neurol 2019; 10:924. [PMID: 31507524 PMCID: PMC6718570 DOI: 10.3389/fneur.2019.00924] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/09/2019] [Indexed: 01/07/2023] Open
Abstract
Background: In recent years pre-clinical stroke research has shown increased interest in the development of biomaterial-based therapies to promote tissue repair and functional recovery. Such strategies utilize biomaterials as structural support for tissue regeneration or as delivery vehicles for therapeutic agents. While a range of biomaterials have been tested in stroke models, currently no overview is available for evaluating the benefit of these approaches. We therefore performed a systematic review and meta-analysis of studies investigating the use of biomaterials for the treatment of stroke in experimental animal models. Methods: Studies were identified by searching electronic databases (PubMed, Web of Science) and reference lists of relevant review articles. Studies reporting lesion volume and/or neurological score were included. Standardized mean difference (SMD) and 95% confidence intervals were calculated using DerSimonian and Laird random effects. Study quality and risk of bias was assessed using the CAMARADES checklist. Publication bias was visualized by funnel plots followed by trim and fill analysis of missing publications. Results: A total of 66 publications were included in the systematic review, of which 44 (86 comparisons) were assessed in the meta-analysis. Overall, biomaterial-based interventions improved both lesion volume (SMD: -2.98, 95% CI: -3.48, -2.48) and neurological score (SMD: -2.3, 95% CI: -2.85, -1.76). The median score on the CAMARADES checklist was 5.5/10 (IQR 4.25-6). Funnel plots of lesion volume and neurological score data revealed pronounced asymmetry and publication bias. Additionally, trim and fill analysis estimated 19 "missing" studies for the lesion volume outcome adjusting the effect size to -1.91 (95% CI: -2.44, -1.38). Conclusions: Biomaterials including scaffolds and particles exerted a positive effect on histological and neurological outcomes in pre-clinical stroke models. However, heterogeneity in the field, publication bias and study quality scores which may be another source of bias call for standardization of outcome measures and improved study reporting.
Collapse
Affiliation(s)
- Faye Bolan
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Irene Louca
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Calvin Heal
- Faculty of Biology, Medicine and Health, Centre for Biostatistics, Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| | - Catriona J. Cunningham
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom,*Correspondence: Catriona J. Cunningham
| |
Collapse
|
90
|
Lim TC, Mandeville E, Weng D, Wang LS, Kurisawa M, Leite-Morris K, Selim MH, Lo EH, Spector M. Hydrogel-Based Therapy for Brain Repair After Intracerebral Hemorrhage. Transl Stroke Res 2019; 11:412-417. [DOI: 10.1007/s12975-019-00721-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 07/04/2019] [Accepted: 07/15/2019] [Indexed: 01/30/2023]
|
91
|
Corey S, Abraham DI, Kaneko Y, Lee JY, Borlongan CV. Selective endovascular cooling for stroke entails brain-derived neurotrophic factor and splenic IL-10 modulation. Brain Res 2019; 1722:146380. [PMID: 31415765 DOI: 10.1016/j.brainres.2019.146380] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/08/2019] [Accepted: 08/12/2019] [Indexed: 12/27/2022]
Abstract
Stroke poses a serious health and economic burden, and the lack of treatment options necessitates a viable therapy. Hypothermia represents a promising stroke therapy, yet side effects of full-body cooling, such as pneumonia, limit its clinical application. Selective endovascular cooling (SEC), via infusion of cold saline through the intraarterial artery, represents an attractive alternative by locally cooling the brain while preserving body temperature. However, the mechanisms underlying SEC are poorly understood. Brain-derived neurotrophic factor (BDNF) is a widely recognized promotor of neuroplasticity and biomarker of stroke outcomes, as well as its association with inflammation, such as IL-10. Stroke-induced neuroinflammation exacerbates damage and stems from peripheral organs, namely the spleen. The spleen has emerged as a therapeutic target for stroke, yet the effect of SEC on the splenic inflammatory response is unknown. Here, we aimed to elucidate the local and peripheral mechanisms driving SEC as a neuroprotective stroke therapy by examining brain BDNF and splenic IL-10 expression. Animals that received SEC prior to stroke displayed elevated brain BDNF expression ipsilaterally and contralaterally across the cortex, striatum, and hippocampus. SEC also upregulated splenic IL-10, suggesting alteration of the peripheral inflammatory response. The oxygen-glucose deprivation in vitro model of stroke further demonstrated that "cold" rat splenocytes protected rat primary neurons by upregulating BDNF and IL-10. Altogether these data support BDNF- and IL-10-based mechanisms underlying the neuroprotective potential of SEC therapy for stroke, and further advance the concept of exploiting the pathological link between brain and spleen as therapeutic targets.
Collapse
Affiliation(s)
- Sydney Corey
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Diego Incontri Abraham
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Yuji Kaneko
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Jea-Young Lee
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA.
| |
Collapse
|
92
|
Zhu Z, Zheng L, Li Y, Huang T, Chao YC, Pan L, Zhu H, Zhao Y, Yu W, Li P. Potential Immunotherapeutic Targets on Myeloid Cells for Neurovascular Repair After Ischemic Stroke. Front Neurosci 2019; 13:758. [PMID: 31447626 PMCID: PMC6696904 DOI: 10.3389/fnins.2019.00758] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/08/2019] [Indexed: 12/11/2022] Open
Abstract
Neurological deficits and cognitive dysfunctions caused by acute ischemic stroke pose enormous burden to the stroke families and the communities. Restoration of the normal function of the neurovascular unit following ischemic stroke is critical for improving neurological recovery and cognitive functions after stroke. Recent evidence suggests that the myeloid cells including both the resident microglia and infiltrating monocytes/macrophages and neutrophils are highly plastic in response to the environmental cues. They intimately interact with multiple components of the neurovascular unit in response to the alarmins, danger associated pattern molecules (DAMPs) and other signals released from the ischemic brain. The aim of this review is to discuss the reciprocal interactions between the myeloid cells and the ischemic neurovascular unit during the late repair phase of cerebral ischemic stroke. We also summarize potential immunotherapeutic targets on myeloid cells and new therapeutic approaches targeting myeloid cells, such as cell transplantation, mitochondrial dynamic and extracellular vesicles-based therapy et al to enhance neurovascular repair for better stroke recovery.
Collapse
Affiliation(s)
- Ziyu Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Li Zheng
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yan Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Tingting Huang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yu-Chieh Chao
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Lijun Pan
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Hui Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yanhua Zhao
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
93
|
Harui A, Roth MD. Employing a glutathione-s-transferase-tag and hyaluronidase to control cytokine retention and release from a hyaluronic acid hydrogel matrix. J Biomater Appl 2019; 34:631-639. [PMID: 31387421 DOI: 10.1177/0885328219867974] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Airi Harui
- David Geffen School of Medicine, Department of Medicine at UCLA, Division of Pulmonary and Critical Care, Los Angeles, CA, USA
| | - Micahel D Roth
- David Geffen School of Medicine, Department of Medicine at UCLA, Division of Pulmonary and Critical Care, Los Angeles, CA, USA
| |
Collapse
|
94
|
Houlton J, Abumaria N, Hinkley SFR, Clarkson AN. Therapeutic Potential of Neurotrophins for Repair After Brain Injury: A Helping Hand From Biomaterials. Front Neurosci 2019; 13:790. [PMID: 31427916 PMCID: PMC6688532 DOI: 10.3389/fnins.2019.00790] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/15/2019] [Indexed: 12/17/2022] Open
Abstract
Stroke remains the leading cause of long-term disability with limited options available to aid in recovery. Significant effort has been made to try and minimize neuronal damage following stroke with use of neuroprotective agents, however, these treatments have yet to show clinical efficacy. Regenerative interventions have since become of huge interest as they provide the potential to restore damaged neural tissue without being limited by a narrow therapeutic window. Neurotrophins, such as brain-derived neurotrophic factor (BDNF), and their high affinity receptors are actively produced throughout the brain and are involved in regulating neuronal activity and normal day-to-day function. Furthermore, neurotrophins are known to play a significant role in both protection and recovery of function following neurodegenerative diseases such as stroke and traumatic brain injury (TBI). Unfortunately, exogenous administration of these neurotrophins is limited by a lack of blood-brain-barrier (BBB) permeability, poor half-life, and rapid degradation. Therefore, we have focused this review on approaches that provide a direct and sustained neurotrophic support using pharmacological therapies and mimetics, physical activity, and potential drug delivery systems, including discussion around advantages and limitations for use of each of these systems. Finally, we discuss future directions of biomaterial drug-delivery systems, including the incorporation of heparan sulfate (HS) in conjunction with neurotrophin-based interventions.
Collapse
Affiliation(s)
- Josh Houlton
- Brain Health Research Centre, Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Nashat Abumaria
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Brain Science, Fudan University, Shanghai, China
- Department of Laboratory Animal Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Simon F. R. Hinkley
- The Ferrier Research Institute, Victoria University of Wellington, Petone, New Zealand
| | - Andrew N. Clarkson
- Brain Health Research Centre, Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
95
|
Influencing neuroplasticity in stroke treatment with advanced biomaterials-based approaches. Adv Drug Deliv Rev 2019; 148:204-218. [PMID: 30579882 DOI: 10.1016/j.addr.2018.12.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 11/05/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023]
Abstract
Since the early 1990s, we have known that the adult brain is not static and has the capacity to repair itself. The delivery of various therapeutic factors and cells have resulted in some exciting pre-clinical and clinical outcomes in stroke models by targeting post-injury plasticity to enhance recovery. Developing a deeper understanding of the pathways that modulate plasticity will enable us to optimize delivery strategies for therapeutics and achieve more robust effects. Biomaterials are a key tool for the optimization of these potential treatments, owing to their biocompatibility and tunability. In this review, we identify factors and targets that impact plastic processes known to contribute to recovery, discuss the role of biomaterials in enhancing the efficacy of treatment strategies, and suggest combinatorial approaches based on the stage of injury progression.
Collapse
|
96
|
|
97
|
Tang-Schomer MD, Kaplan DL, Whalen MJ. Film interface for drug testing for delivery to cells in culture and in the brain. Acta Biomater 2019; 94:306-319. [PMID: 30836199 DOI: 10.1016/j.actbio.2019.02.052] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 12/31/2022]
Abstract
Brain access remains a major challenge in drug testing. The nearly 'impermeable' blood-brain-barrier (BBB) prevents most drugs from gaining access to brain cells via systematic intravenous (IV) injection. In this study, silk fibroin films were used as drug carrier as well as cell culture substrate to simulate the in vivo interface between drug reservoir and brain cells for testing drug delivery in the brain. In in vitro studies, film-released arabinofuranosyl cytidine (AraC), a mitotic inhibitor, selectively killed glial cells in film-supported mixed neural cell cultures; with widened dosage windows for drug efficacy and tolerance compared to drugs in solution. In the brain, the presence of silk films was well tolerated with no signs of acute neuroinflammation, cell death, or altered brain function. Topical application of silk films on the cortical surface delivered Evans blue, a BBB-impenetrable fluorescent marker, through the intact dura matter into the parenchyma of the ipsilateral hemisphere as deep as the hippocampal region, but not the contralateral hemisphere. In a mouse traumatic brain injury (TBI) model, necrosis markers by film delivery accessed more cells in the lesion core than by con-current IV delivery; whereas the total coverage including the peri-lesional area appeared to be comparable between the two routes. The complementary distribution patterns of co-delivered markers provided direct evidence of the partial confinement of either route's access to brain cells by a restrictive zone near the lesion border. Finally, film-delivered necrostatin-1 reduced overall cell necrosis by approximately 40% in the TBI model. These findings from representative small molecules of delivery route-dependent drug access are broadly applicable for evaluating drug actions both in vitro and in vivo. Combined with its demonstrated role of supporting neuron-electrode interfaces, the film system can be further developed for testing a range of neuromodulation approaches (i.e., drug delivery, electrical stimulation, cell graft) in the brain. STATEMENT OF SIGNIFICANCE: This study demonstrated that silk fibroin films can be used to evaluate drug actions both in vitro and in vivo, partially overcoming the significant delivery barriers of the brain. This system can be adapted for efficient drug access to specific brain regions and/or cell types. The film system can be further developed for testing a range of interventions with drugs, electrical signals or cell graft for analysis of treatment outcomes including cell responses and brain function.
Collapse
Affiliation(s)
- Min D Tang-Schomer
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; University of Connecticut Health Center & Connecticut Children's Medical Center, Department of Pediatrics, Farmington, CT 06032, USA.
| | - David L Kaplan
- Tufts University, Department of Biomedical Engineering, Medford, MA 02155, United States.
| | - Michael J Whalen
- Harvard Medical School, Acute Brain Injury Research Laboratory, Massachusetts General Hospital for Children, Charlestown, MA 02129, United States.
| |
Collapse
|
98
|
Obermeyer JM, Tuladhar A, Payne SL, Ho E, Morshead CM, Shoichet MS. Local Delivery of Brain-Derived Neurotrophic Factor Enables Behavioral Recovery and Tissue Repair in Stroke-Injured Rats. Tissue Eng Part A 2019; 25:1175-1187. [DOI: 10.1089/ten.tea.2018.0215] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Jaclyn M. Obermeyer
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
| | - Anup Tuladhar
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
| | - Samantha L. Payne
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
| | - Eric Ho
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
| | - Cindi M. Morshead
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
- Department of Surgery, University of Toronto, Toronto, Canada
| | - Molly S. Shoichet
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
- Department of Chemistry, University of Toronto, Toronto, Canada
| |
Collapse
|
99
|
Elzib H, Pawloski J, Ding Y, Asmaro K. Antidepressant pharmacotherapy and poststroke motor rehabilitation: A review of neurophysiologic mechanisms and clinical relevance. Brain Circ 2019; 5:62-67. [PMID: 31334358 PMCID: PMC6611192 DOI: 10.4103/bc.bc_3_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/06/2019] [Accepted: 04/04/2019] [Indexed: 12/23/2022] Open
Abstract
According to the National Stroke Association, stroke is the leading cause of adult disability in the United States, where it is estimated that about 795,000 strokes occur on an annual basis. Minimizing the disability burden of a stroke routinely involves behavioral therapies such as physical and occupational therapy, as well as pharmacologic interventions. The positive effect of antidepressants on functional outcomes for patients with poststroke depression is well known and practiced. In the past 15 years, a growing body of evidence has demonstrated that antidepressant pharmacotherapy and selective serotonin reuptake inhibitors specifically have a role in the functional recovery from strokes even in the nondepressed population. The mechanisms by which antidepressants improve motor recovery following stroke are multifactorial, but it is clear that the process involves augmentation of cerebral blood flow, cortical excitation, and potentiation of neural growth factors all resulting in enhancement of neurogeneration. This review will examine the existing evidence and mechanisms behind antidepressant use for motor recovery in stroke patients and discuss the major human clinical trials that have been conducted surrounding this topic. The evidence clearly suggests that antidepressants have a positive impact on poststroke functional recovery regardless of the presence of depression, and although large-scale randomized, controlled trials are still ongoing, antidepressants are emerging as a promising pharmaceutical means of actively lessening the burden of disability following stroke.
Collapse
Affiliation(s)
- Haya Elzib
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Jacob Pawloski
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Neurosurgery, Henry Ford Health System, Detroit, Michigan, USA
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Karam Asmaro
- Department of Neurosurgery, Henry Ford Health System, Detroit, Michigan, USA
| |
Collapse
|
100
|
Modo M, Badylak SF. A roadmap for promoting endogenous in situ tissue restoration using inductive bioscaffolds after acute brain injury. Brain Res Bull 2019; 150:136-149. [PMID: 31128250 DOI: 10.1016/j.brainresbull.2019.05.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 05/10/2019] [Accepted: 05/17/2019] [Indexed: 02/08/2023]
Abstract
The regeneration of brain tissue remains one of the greatest unsolved challenges in medicine and by many is considered unfeasible. Indeed, the adult mammalian brain does not regenerate tissue, but there is ongoing endogenous neurogenesis, which is upregulated after injury and contributes to tissue repair. This endogenous repair response is a conditio sine que non for tissue regeneration. However, scarring around the lesion core and cavitation provide unfavorable conditions for tissue regeneration in the brain. Based on the success of using extracellular matrix (ECM)-based bioscaffolds in peripheral soft tissue regeneration, it is plausible that the provision of an inductive ECM-based hydrogel inside the volumetric tissue loss can attract neural cells and create a de novo viable tissue. Following perturbation theory of these successes in peripheral tissues, we here propose 9 perturbation parts (i.e. requirements) that can be solved independently to create an integrated series to build a functional and integrated de novo neural tissue. Necessities for tissue formation, anatomical and functional connectivity are further discussed to provide a new substrate to support the improvement of behavioral impairments after acute brain injury. We also consider potential parallel developments of this tissue engineering effort that can support therapeutic benefits in the absence of de novo tissue formation (e.g. structural support to veterate brain tissue). It is envisaged that eventually top-down inductive "natural" bioscaffolds composed of decellularized tissues (i.e. ECM) will be replaced by bottom-up synthetic designer hydrogels that will provide very defined structural and signaling properties, potentially even opening up opportunities we currently do not envisage using natural materials.
Collapse
Affiliation(s)
- Michel Modo
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, USA; University of Pittsburgh, Department of Bioengineering, Pittsburgh, PA, USA; University of Pittsburgh, Department of Radiology, Pittsburgh, PA, USA.
| | - Stephen F Badylak
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, USA; University of Pittsburgh, Department of Bioengineering, Pittsburgh, PA, USA; University of Pittsburgh, Department of Surgery, Pittsburgh, PA, USA
| |
Collapse
|