1
|
Jinawong K, Piamsiri C, Apaijai N, Maneechote C, Arunsak B, Nawara W, Thonusin C, Pintana H, Chattipakorn N, Chattipakorn SC. Modulating Mitochondrial Dynamics Mitigates Cognitive Impairment in Rats with Myocardial Infarction. Curr Neuropharmacol 2024; 22:1749-1760. [PMID: 38362882 PMCID: PMC11284718 DOI: 10.2174/1570159x22666240131114913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND We have previously demonstrated that oxidative stress and brain mitochondrial dysfunction are key mediators of brain pathology during myocardial infarction (MI). OBJECTIVE To investigate the beneficial effects of mitochondrial dynamic modulators, including mitochondrial fission inhibitor (Mdivi-1) and mitochondrial fusion promotor (M1), on cognitive function and molecular signaling in the brain of MI rats in comparison with the effect of enalapril. METHODS Male rats were assigned to either sham or MI operation. In the MI group, rats with an ejection Fraction less than 50% were included, and then they received one of the following treatments for 5 weeks: vehicle, enalapril, Mdivi-1, or M1. Cognitive function was tested, and the brains were used for molecular study. RESULTS MI rats exhibited cardiac dysfunction with systemic oxidative stress. Cognitive impairment was found in MI rats, along with dendritic spine loss, blood-brain barrier (BBB) breakdown, brain mitochondrial dysfunction, and decreased mitochondrial and increased glycolysis metabolism, without the alteration of APP, BACE-1, Tau and p-Tau proteins. Treatment with Mdivi-1, M1, and enalapril equally improved cognitive function in MI rats. All treatments decreased dendritic spine loss, brain mitochondrial oxidative stress, and restored mitochondrial metabolism. Brain mitochondrial fusion was recovered only in the Mdivi-1-treated group. CONCLUSION Mitochondrial dynamics modulators improved cognitive function in MI rats through a reduction of systemic oxidative stress and brain mitochondrial dysfunction and the enhancement of mitochondrial metabolism. In addition, this mitochondrial fission inhibitor increased mitochondrial fusion in MI rats.
Collapse
Affiliation(s)
- Kewarin Jinawong
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chanon Piamsiri
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nattayaporn Apaijai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chayodom Maneechote
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Busarin Arunsak
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wichwara Nawara
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chanisa Thonusin
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Hiranya Pintana
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C. Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
2
|
Huang W, Qiu W, Chen K, Ye S, Wang D, Hu J, Xu H, Lin L, Li X. Research progress of fibroblast growth factor in nervous system diseases. Zhejiang Da Xue Xue Bao Yi Xue Ban 2022; 51:738-749. [PMID: 36915973 PMCID: PMC10262007 DOI: 10.3724/zdxbyxb-2022-0180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/25/2022] [Indexed: 06/17/2023]
Abstract
Fibroblast growth factors (FGF) are a group of structurally related polypeptides which constitute an elaborate signaling system with their receptors. Evidence accumulated in the years suggests that the FGF family plays a key role in the repair of central nervous system injury. The main protective mechanisms include activating the expression of PI3K-Akt, peroxisome proliferator-activated receptor (PPARγ) and other signals; inhibiting NF-κB-mediated inflammatory response, oxidative stress and apoptosis; regulating neuronal differentiation and neuronal excitability as well as participating in protection of neurovascular units and nerve function repair. This paper comprehensively summarizes the latest research progress in FGF signaling related to diseases of the central nervous system such as cerebral infarction, cerebral hemorrhage, traumatic brain injury, Alzheimer's disease, Parkinson's disease, epilepsy and depression, aiming to provide scientific basis and reference for the development of innovative FGF drugs for the prevention and treatment of neurological diseases.
Collapse
Affiliation(s)
- Wenting Huang
- 1. Department of Neurology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Wanhua Qiu
- 2. School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Kun Chen
- 2. School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Shasha Ye
- 2. School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Dongxue Wang
- 2. School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Jian Hu
- 2. School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Huiqin Xu
- 1. Department of Neurology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Li Lin
- 2. School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Xiaokun Li
- 2. School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| |
Collapse
|
3
|
An implantable human stem cell-derived tissue-engineered rostral migratory stream for directed neuronal replacement. Commun Biol 2021; 4:879. [PMID: 34267315 PMCID: PMC8282659 DOI: 10.1038/s42003-021-02392-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 06/15/2021] [Indexed: 11/16/2022] Open
Abstract
The rostral migratory stream (RMS) facilitates neuroblast migration from the subventricular zone to the olfactory bulb throughout adulthood. Brain lesions attract neuroblast migration out of the RMS, but resultant regeneration is insufficient. Increasing neuroblast migration into lesions has improved recovery in rodent studies. We previously developed techniques for fabricating an astrocyte-based Tissue-Engineered RMS (TE-RMS) intended to redirect endogenous neuroblasts into distal brain lesions for sustained neuronal replacement. Here, we demonstrate that astrocyte-like-cells can be derived from adult human gingiva mesenchymal stem cells and used for TE-RMS fabrication. We report that key proteins enriched in the RMS are enriched in TE-RMSs. Furthermore, the human TE-RMS facilitates directed migration of immature neurons in vitro. Finally, human TE-RMSs implanted in athymic rat brains redirect migration of neuroblasts out of the endogenous RMS. By emulating the brain’s most efficient means for directing neuroblast migration, the TE-RMS offers a promising new approach to neuroregenerative medicine. O’Donnell et al. describe their Tissue-Engineered Rostral Migratory Stream (TE-RMS) comprised of human astrocyte-like cells that can be derived from adult gingival stem cells within one week, which reorganizes into bundles of bidirectional, longitudinally-aligned astrocytes to emulate the endogenous RMS. Establishing immature neuronal migration in vitro and in vivo, their study demonstrates surgical feasibility and proof-of-concept evidence for this nascent technology.
Collapse
|
4
|
Park E, Lyon JG, Alvarado-Velez M, Betancur MI, Mokarram N, Shin JH, Bellamkonda RV. Enriching neural stem cell and anti-inflammatory glial phenotypes with electrical stimulation after traumatic brain injury in male rats. J Neurosci Res 2021; 99:1864-1884. [PMID: 33772860 PMCID: PMC8360147 DOI: 10.1002/jnr.24834] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/08/2021] [Indexed: 12/26/2022]
Abstract
Traumatic brain injury (TBI) by an external physical impact results in compromised brain function via undesired neuronal death. Following the injury, resident and peripheral immune cells, astrocytes, and neural stem cells (NSCs) cooperatively contribute to the recovery of the neuronal function after TBI. However, excessive pro‐inflammatory responses of immune cells, and the disappearance of endogenous NSCs at the injury site during the acute phase of TBI, can exacerbate TBI progression leading to incomplete healing. Therefore, positive outcomes may depend on early interventions to control the injury‐associated cellular milieu in the early phase of injury. Here, we explore electrical stimulation (ES) of the injury site in a rodent model (male Sprague–Dawley rats) to investigate its overall effect on the constituent brain cell phenotype and composition during the acute phase of TBI. Our data showed that a brief ES for 1 hr on day 2 of TBI promoted anti‐inflammatory phenotypes of microglia as assessed by CD206 expression and increased the population of NSCs and Nestin+ astrocytes at 7 days post‐TBI. Also, ES effectively increased the number of viable neurons when compared to the unstimulated control group. Given the salience of microglia and neural stem cells for healing after TBI, our results strongly support the potential benefit of the therapeutic use of ES during the acute phase of TBI to regulate neuroinflammation and to enhance neuroregeneration.
Collapse
Affiliation(s)
- Eunyoung Park
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Johnathan G Lyon
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Melissa Alvarado-Velez
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Martha I Betancur
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Nassir Mokarram
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Jennifer H Shin
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Ravi V Bellamkonda
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
5
|
Iwasa SN, Shi HH, Hong SH, Chen T, Marquez-Chin M, Iorio-Morin C, Kalia SK, Popovic MR, Naguib HE, Morshead CM. Novel Electrode Designs for Neurostimulation in Regenerative Medicine: Activation of Stem Cells. Bioelectricity 2020; 2:348-361. [PMID: 34471854 PMCID: PMC8370381 DOI: 10.1089/bioe.2020.0034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neural stem and progenitor cells (i.e., neural precursors) are found within specific regions in the central nervous system and have great regenerative capacity. These cells are electrosensitive and their behavior can be regulated by the presence of electric fields (EFs). Electrical stimulation is currently used to treat neurological disorders in a clinical setting. Herein we propose that electrical stimulation can be used to enhance neural repair by regulating neural precursor cell (NPC) kinetics and promoting their migration to sites of injury or disease. We discuss how intrinsic and extrinsic factors can affect NPC migration in the presence of an EF and how this impacts electrode design with the goal of enhancing tissue regeneration. We conclude with an outlook on future clinical applications of electrical stimulation and highlight technological advances that would greatly support these applications.
Collapse
Affiliation(s)
- Stephanie N Iwasa
- The KITE Research Institute, Toronto Rehabilitation Institute-University Health Network, Toronto, Canada
- CRANIA, University Health Network and University of Toronto, Toronto, Canada
| | - HaoTian H Shi
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Canada
| | - Sung Hwa Hong
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Canada
| | - Tianhao Chen
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Melissa Marquez-Chin
- The KITE Research Institute, Toronto Rehabilitation Institute-University Health Network, Toronto, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Christian Iorio-Morin
- Department of Neurosurgery, University Health Network, University of Toronto, Toronto, Canada
| | - Suneil K Kalia
- The KITE Research Institute, Toronto Rehabilitation Institute-University Health Network, Toronto, Canada
- CRANIA, University Health Network and University of Toronto, Toronto, Canada
- Department of Neurosurgery, University Health Network, University of Toronto, Toronto, Canada
- Krembil Research Institute, Toronto, Canada
| | - Milos R Popovic
- The KITE Research Institute, Toronto Rehabilitation Institute-University Health Network, Toronto, Canada
- CRANIA, University Health Network and University of Toronto, Toronto, Canada
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Hani E Naguib
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
- Department of Materials Science & Engineering, University of Toronto, Toronto, Canada
| | - Cindi M Morshead
- The KITE Research Institute, Toronto Rehabilitation Institute-University Health Network, Toronto, Canada
- CRANIA, University Health Network and University of Toronto, Toronto, Canada
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
- Department of Surgery, University of Toronto, Toronto, Canada
| |
Collapse
|
6
|
Menet R, Lecordier S, ElAli A. Wnt Pathway: An Emerging Player in Vascular and Traumatic Mediated Brain Injuries. Front Physiol 2020; 11:565667. [PMID: 33071819 PMCID: PMC7530281 DOI: 10.3389/fphys.2020.565667] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022] Open
Abstract
The Wnt pathway, which comprises the canonical and non-canonical pathways, is an evolutionarily conserved mechanism that regulates crucial biological aspects throughout the development and adulthood. Emergence and patterning of the nervous and vascular systems are intimately coordinated, a process in which Wnt pathway plays particularly important roles. In the brain, Wnt ligands activate a cell-specific surface receptor complex to induce intracellular signaling cascades regulating neurogenesis, synaptogenesis, neuronal plasticity, synaptic plasticity, angiogenesis, vascular stabilization, and inflammation. The Wnt pathway is tightly regulated in the adult brain to maintain neurovascular functions. Historically, research in neuroscience has emphasized essentially on investigating the pathway in neurodegenerative disorders. Nonetheless, emerging findings have demonstrated that the pathway is deregulated in vascular- and traumatic-mediated brain injuries. These findings are suggesting that the pathway constitutes a promising target for the development of novel therapeutic protective and restorative interventions. Yet, targeting a complex multifunctional signal transduction pathway remains a major challenge. The review aims to summarize the current knowledge regarding the implication of Wnt pathway in the pathobiology of ischemic and hemorrhagic stroke, as well as traumatic brain injury (TBI). Furthermore, the review will present the strategies used so far to manipulate the pathway for therapeutic purposes as to highlight potential future directions.
Collapse
Affiliation(s)
- Romain Menet
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Sarah Lecordier
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Ayman ElAli
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
7
|
Pudełek M, Król K, Catapano J, Wróbel T, Czyż J, Ryszawy D. Epidermal Growth Factor (EGF) Augments the Invasive Potential of Human Glioblastoma Multiforme Cells via the Activation of Collaborative EGFR/ROS-Dependent Signaling. Int J Mol Sci 2020; 21:ijms21103605. [PMID: 32443749 PMCID: PMC7279139 DOI: 10.3390/ijms21103605] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 01/06/2023] Open
Abstract
Abnormal secretion of epidermal growth factor (EGF) by non-neuronal cells (e.g., glioma-associated microglia) establishes a feedback loop between glioblastoma multiforme (GBM) invasion and a functional disruption of brain tissue. Considering the postulated significance of this vicious circle for GBM progression, we scrutinized mechanisms of EGF-dependent pro-invasive signaling in terms of its interrelations with energy metabolism and reactive oxygen species (ROS) production. The effects of EGF on the invasiveness of human glioblastoma T98G cells were estimated using time-lapse video microscopy, immunocytochemistry, cell cycle assay, immunoblot analyses, and Transwell® assay. These techniques were followed by quantification of the effect of EGFR (Epidermal Growth Factor Receptor) and ROS inhibitors on the EGF-induced T98G invasiveness and intracellular ROS, ATP, and lactate levels and mitochondrial metabolism. The EGF remarkably augmented the proliferation and motility of the T98G cells. Responses of these cells were accompanied by cellular rear–front polarization, translocation of vinculin to the leading lamellae, and increased promptness of penetration of micropore barriers. Erlotinib (the EGFR inhibitor) significantly attenuated the EGF-induced T98G invasiveness and metabolic reprogramming of the T98G cells, otherwise illustrated by the increased mitochondrial activity, glycolysis, and ROS production in the EGF-treated cells. In turn, ROS inhibition by N-acetyl-L-cysteine (NAC) had no effect on T98G morphology, but considerably attenuated EGF-induced cell motility. Our data confirmed the EGFR/ROS-dependent pro-neoplastic and pro-invasive activity of EGF in human GBM. These EGF effects may depend on metabolic reprogramming of GBM cells and are executed by alternative ROS-dependent/-independent pathways. The EGF may thus preserve bioenergetic homeostasis of GBM cells in hypoxic regions of brain tissue.
Collapse
|
8
|
Sun MK, Passaro AP, Latchoumane CF, Spellicy SE, Bowler M, Goeden M, Martin WJ, Holmes PV, Stice SL, Karumbaiah L. Extracellular Vesicles Mediate Neuroprotection and Functional Recovery after Traumatic Brain Injury. J Neurotrauma 2020; 37:1358-1369. [PMID: 31774030 DOI: 10.1089/neu.2019.6443] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The lack of effective therapies for moderate-to-severe traumatic brain injuries (TBIs) leaves patients with lifelong disabilities. Neural stem cells (NSCs) have demonstrated great promise for neural repair and regeneration. However, direct evidence to support their use as a cell replacement therapy for neural injuries is currently lacking. We hypothesized that NSC-derived extracellular vesicles (NSC EVs) mediate repair indirectly after TBI by enhancing neuroprotection and therapeutic efficacy of endogenous NSCs. We evaluated the short-term effects of acute intravenous injections of NSC EVs immediately following a rat TBI. Male NSC EV-treated rats demonstrated significantly reduced lesion sizes, enhanced presence of endogenous NSCs, and attenuated motor function impairments 4 weeks post-TBI, when compared with vehicle- and TBI-only male controls. Although statistically not significant, we observed a therapeutic effect of NSC EVs on brain lesion volume, nestin expression, and behavioral recovery in female subjects. Our study demonstrates the neuroprotective and functional benefits of NSC EVs for treating TBI and points to gender-dependent effects on treatment outcomes, which requires further investigation.
Collapse
Affiliation(s)
- Min Kyoung Sun
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
- Interdisciplinary Neuroscience Program, University of Georgia, Athens, Georgia, USA
| | - Austin P Passaro
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
- Interdisciplinary Neuroscience Program, University of Georgia, Athens, Georgia, USA
| | - Charles-Francois Latchoumane
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
- Department of Animal and Dairy Science, University of Georgia, Athens, Georgia, USA
| | - Samantha E Spellicy
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
- Interdisciplinary Neuroscience Program, University of Georgia, Athens, Georgia, USA
| | - Michael Bowler
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Morgan Goeden
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - William J Martin
- Animal Health Research Center, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Philip V Holmes
- Interdisciplinary Neuroscience Program, University of Georgia, Athens, Georgia, USA
- Department of Psychology, University of Georgia, Athens, Georgia, USA
| | - Steven L Stice
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
- Interdisciplinary Neuroscience Program, University of Georgia, Athens, Georgia, USA
- Department of Animal and Dairy Science, University of Georgia, Athens, Georgia, USA
| | - Lohitash Karumbaiah
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
- Interdisciplinary Neuroscience Program, University of Georgia, Athens, Georgia, USA
- Department of Animal and Dairy Science, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
9
|
Zambusi A, Ninkovic J. Regeneration of the central nervous system-principles from brain regeneration in adult zebrafish. World J Stem Cells 2020; 12:8-24. [PMID: 32110272 PMCID: PMC7031763 DOI: 10.4252/wjsc.v12.i1.8] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/25/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023] Open
Abstract
Poor recovery of neuronal functions is one of the most common healthcare challenges for patients with different types of brain injuries and/or neurodegenerative diseases. Therapeutic interventions face two major challenges: (1) How to generate neurons de novo to replenish the neuronal loss caused by injuries or neurodegeneration (restorative neurogenesis) and (2) How to prevent or limit the secondary tissue damage caused by long-term accumulation of glial cells, including microglia, at injury site (glial scar). In contrast to mammals, zebrafish have extensive regenerative capacity in numerous vital organs, including the brain, thus making them a valuable model to improve the existing therapeutic approaches for human brain repair. In response to injuries to the central nervous system (CNS), zebrafish have developed specific mechanisms to promote the recovery of the lost tissue architecture and functionality of the damaged CNS. These mechanisms include the activation of a restorative neurogenic program in a specific set of glial cells (ependymoglia) and the resolution of both the glial scar and inflammation, thus enabling proper neuronal specification and survival. In this review, we discuss the cellular and molecular mechanisms underlying the regenerative ability in the adult zebrafish brain and conclude with the potential applicability of these mechanisms in repair of the mammalian CNS.
Collapse
Affiliation(s)
- Alessandro Zambusi
- Helmholtz Center Munich, Biomedical Center, Inst Stem Cell Res, Institute of Stem Cell Research, Department of Cell Biology and Anatomy, University of Munich, Planegg 82152, Germany
| | - Jovica Ninkovic
- Helmholtz Center Munich, Biomedical Center, Inst Stem Cell Res, Institute of Stem Cell Research, Department of Cell Biology and Anatomy, University of Munich, Planegg 82152, Germany
| |
Collapse
|
10
|
Boone DR, Weisz HA, Willey HE, Torres KEO, Falduto MT, Sinha M, Spratt H, Bolding IJ, Johnson KM, Parsley MA, DeWitt DS, Prough DS, Hellmich HL. Traumatic brain injury induces long-lasting changes in immune and regenerative signaling. PLoS One 2019; 14:e0214741. [PMID: 30943276 PMCID: PMC6447179 DOI: 10.1371/journal.pone.0214741] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 03/19/2019] [Indexed: 12/19/2022] Open
Abstract
There are no existing treatments for the long-term degenerative effects of traumatic brain injury (TBI). This is due, in part, to our limited understanding of chronic TBI and uncertainty about which proposed mechanisms for long-term neurodegeneration are amenable to treatment with existing or novel drugs. Here, we used microarray and pathway analyses to interrogate TBI-induced gene expression in the rat hippocampus and cortex at several acute, subchronic and chronic intervals (24 hours, 2 weeks, 1, 2, 3, 6 and 12 months) after parasagittal fluid percussion injury. We used Ingenuity pathway analysis (IPA) and Gene Ontology enrichment analysis to identify significantly expressed genes and prominent cell signaling pathways that are dysregulated weeks to months after TBI and potentially amenable to therapeutic modulation. We noted long-term, coordinated changes in expression of genes belonging to canonical pathways associated with the innate immune response (i.e., NF-κB signaling, NFAT signaling, Complement System, Acute Phase Response, Toll-like receptor signaling, and Neuroinflammatory signaling). Bioinformatic analysis suggested that dysregulation of these immune mediators—many are key hub genes—would compromise multiple cell signaling pathways essential for homeostatic brain function, particularly those involved in cell survival and neuroplasticity. Importantly, the temporal profile of beneficial and maladaptive immunoregulatory genes in the weeks to months after the initial TBI suggests wider therapeutic windows than previously indicated.
Collapse
Affiliation(s)
- Deborah R. Boone
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Harris A. Weisz
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Hannah E. Willey
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | | | - Michael T. Falduto
- GenUs Biosystems, Northbrook, Illinois, United States of America
- Paradise Genomics, Inc., Northbrook, Illinois, United States of America
| | - Mala Sinha
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Heidi Spratt
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Ian J. Bolding
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kathea M. Johnson
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Margaret A. Parsley
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Douglas S. DeWitt
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Donald S. Prough
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Helen L. Hellmich
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
11
|
Dewan S, Schimmel S, Borlongan CV. Treating childhood traumatic brain injury with autologous stem cell therapy. Expert Opin Biol Ther 2018; 18:515-524. [PMID: 29421958 PMCID: PMC6086119 DOI: 10.1080/14712598.2018.1439473] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Neonatal traumatic brain injury (TBI) is a significant cause of developmental disorders. Autologous stem cell therapy may enhance neonatal brain plasticity towards repair of the injured neonatal brain. AREAS COVERED The endogenous neonatal anti-inflammatory response can be enhanced through the delivery of anti-inflammatory agents. Stem cell therapy stands as a robust approach for sequestering the inflammation-induced cell death in the injured brain. Here, we discuss the use of umbilical cord blood cells and bone marrow stromal cells for acute and chronic treatment of experimental neonatal TBI. Autologous stem cell transplantation may dampen neuroinflammation. Clinical translation of this stem cell therapy will require identifying the therapeutic window post-injury and harvesting ample supply of transplantable autologous stem cells. Stem cell banking of cryopreserved cells may allow readily available transplantable cells and circumvent the unpredictable nature of neonatal TBI. Harnessing the anti-inflammatory properties of stem cells is key in combating the progressive neurodegeneration after the initial injury. EXPERT OPINION Combination treatments, such as with hypothermia, may enhance the therapeutic effects of stem cells. Stem cell therapy has immense potential as a stand-alone or adjunctive therapy for treating neuroinflammation associated with neonatal TBI acutely and for preventing further progression of the injury.
Collapse
Affiliation(s)
- Shyam Dewan
- Center of Excellence for Aging and Brain Repair, Department of Neurosugery and Brain Repair, University of South Florida Morsani College of Medicine. 3515 E. Fletcher Avenue, Tampa, FL 33613, USA
| | - Samantha Schimmel
- Center of Excellence for Aging and Brain Repair, Department of Neurosugery and Brain Repair, University of South Florida Morsani College of Medicine. 3515 E. Fletcher Avenue, Tampa, FL 33613, USA
| | - Cesar V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosugery and Brain Repair, University of South Florida Morsani College of Medicine. 3515 E. Fletcher Avenue, Tampa, FL 33613, USA
| |
Collapse
|
12
|
Jia J, Chen F, Wu Y. Recombinant PEP-1-SOD1 improves functional recovery after neural stem cell transplantation in rats with traumatic brain injury. Exp Ther Med 2018; 15:2929-2935. [PMID: 29599832 PMCID: PMC5867477 DOI: 10.3892/etm.2018.5781] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/09/2016] [Indexed: 12/25/2022] Open
Abstract
The transplantation of neural stem cells (NSCs) has been demonstrated as a potential treatment strategy for traumatic brain injury (TBI). Cu, Zn-superoxide dismutase (SOD1) is an important antioxidant enzyme that detoxifies intracellular reactive oxygen species, thereby protecting cells from oxidative damage. PEP-1, a peptide carrier, is able to deliver full-length native peptides or proteins into cells. Therefore, the current study investigated the effect of the transplantation of NSCs in combination with PEP-1-SOD1 for the treatment of experimental TBI in rats. Initially, the effect of PEP-1-SOD1 on the proliferation of NSCs was evaluated by MTT assay. PEP-1-SOD1 (0.5, 2.5 and 4.5 µM) significantly increased the proliferation rates of NSCs at 24, 48 and 72 h in a dose-dependent manner. PEP-1-SOD1 also promoted the differentiation of NSCs in vitro. The in vivo experiment showed that PEP-1-SOD1 in combination with NSC transplantation significantly improved the functional recovery of rats following TBI compared with NSC transplantation alone. A significant increase in brain aquaporin-4 (AQP4) mRNA and protein expression levels was observed 4 days post-TBI in PEP-1-SOD1, NSCs and PEP-1-SOD1 + NSCs groups compared with the saline group. The PEP-1-SOD1 + NSCs group showed a further increase of AQP4 mRNA and protein expression levels compared with the NSCs and PEP-1-SOD1 groups. In conclusion, the current data suggests that PEP-1-SOD1 may promote the proliferation and differentiation of NSCs, and thereby improve the functional recovery of TBI model rats following NSCs transplantation through upregulating the expression of AQP4.
Collapse
Affiliation(s)
- Jinming Jia
- Department of Critical Care Medicine, The Putian Hanjiang Hospital, Putian, Fujian 351100, P.R. China
| | - Feifei Chen
- Department of Emergency, The Third People's Hospital of Changzhou, Changzhou, Jiangsu 213001, P.R. China
| | - Yunfei Wu
- Department of Pathology, The Third People's Hospital of Changzhou, Changzhou, Jiangsu 213001, P.R. China
| |
Collapse
|
13
|
Lecca D, Fumagalli M, Ceruti S, Abbracchio MP. Intertwining extracellular nucleotides and their receptors with Ca2+ in determining adult neural stem cell survival, proliferation and final fate. Philos Trans R Soc Lond B Biol Sci 2017; 371:rstb.2015.0433. [PMID: 27377726 DOI: 10.1098/rstb.2015.0433] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2016] [Indexed: 02/07/2023] Open
Abstract
In the central nervous system (CNS), during both brain and spinal cord development, purinergic and pyrimidinergic signalling molecules (ATP, UTP and adenosine) act synergistically with peptidic growth factors in regulating the synchronized proliferation and final specification of multipotent neural stem cells (NSCs) to neurons, astrocytes or oligodendrocytes, the myelin-forming cells. Some NSCs still persist throughout adulthood in both specific 'neurogenic' areas and in brain and spinal cord parenchyma, retaining the potentiality to generate all the three main types of adult CNS cells. Once CNS anatomical structures are defined, purinergic molecules participate in calcium-dependent neuron-to-glia communication and also control the behaviour of adult NSCs. After development, some purinergic mechanisms are silenced, but can be resumed after injury, suggesting a role for purinergic signalling in regeneration and self-repair also via the reactivation of adult NSCs. In this respect, at least three different types of adult NSCs participate in the response of the adult brain and spinal cord to insults: stem-like cells residing in classical neurogenic niches, in particular, in the ventricular-subventricular zone (V-SVZ), parenchymal oligodendrocyte precursor cells (OPCs, also known as NG2-glia) and parenchymal injury-activated astrocytes (reactive astrocytes). Here, we shall review and discuss the purinergic regulation of these three main adult NSCs, with particular focus on how and to what extent modulation of intracellular calcium levels by purinoceptors is mandatory to determine their survival, proliferation and final fate.This article is part of the themed issue 'Evolution brings Ca(2+) and ATP together to control life and death'.
Collapse
Affiliation(s)
- Davide Lecca
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy
| | - Marta Fumagalli
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy
| | - Stefania Ceruti
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy
| | - Maria P Abbracchio
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy
| |
Collapse
|
14
|
Dutta D, Hickey K, Salifu M, Fauer C, Willingham C, Stabenfeldt SE. Spatiotemporal presentation of exogenous SDF-1 with PLGA nanoparticles modulates SDF-1/CXCR4 signaling axis in the rodent cortex. Biomater Sci 2017; 5:1640-1651. [PMID: 28703822 PMCID: PMC5588897 DOI: 10.1039/c7bm00489c] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Stromal cell-derived factor-1 (SDF-1) and its key receptor CXCR4 have been implicated in directing cellular recruitment for several pathological/disease conditions thus also gained considerable attention for regenerative medicine. One regenerative approach includes sustained release of SDF-1 to stimulate prolonged stem cell recruitment. However, the impact of SDF-1 sustained release on the endogenous SDF-1/CXCR4 signaling axis is largely unknown as auto-regulatory mechanisms typically dictate cytokine/receptor signaling. We hypothesize that spatiotemporal presentation of exogenous SDF-1 is a key factor in achieving long-term manipulation of endogenous SDF-1/CXCR4 signaling. Here in the present study, we sought to probe our hypothesis using a transgenic mouse model to contrast the spatial activation of endogenous SDF-1 and CXCR4 in response to exogenous SDF-1 injected in bolus or controlled release (PLGA nanoparticles) form in the adult rodent cortex. Our data suggests that the manner of SDF-1 presentation significantly affected initial CXCR4 cellular activation/recruitment despite having similar protein payloads over the first 24 h (∼30 ng for both bolus and sustained release groups). Yet, one week post-injection, this response was negligible. Therefore, the transient nature CXCR4 recruitment/activation in response to bolus or controlled release SDF-1 indicated that cytokine/receptor auto-regulatory mechanisms may demand more complex release profiles (i.e. delayed and/or pulsed release) to achieve sustained cellular response.
Collapse
Affiliation(s)
- D Dutta
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| | - K Hickey
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| | - M Salifu
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| | - C Fauer
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| | - C Willingham
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| | - S E Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
15
|
Kim C, Park JM, Kong T, Lee S, Seo KW, Choi Y, Song YS, Moon J. Double-Injected Human Stem Cells Enhance Rehabilitation in TBI Mice Via Modulation of Survival and Inflammation. Mol Neurobiol 2017; 55:4870-4884. [PMID: 28736792 PMCID: PMC5948256 DOI: 10.1007/s12035-017-0683-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 07/07/2017] [Indexed: 12/22/2022]
Abstract
Traumatic brain injury (TBI), a complicated form of brain damage, is a major cause of mortality in adults. Following mechanical and structural primary insults, a battery of secondary insults, including neurotransmitter-mediated cytotoxicity, dysregulation of calcium and macromolecule homeostasis, and increased oxidative stress, exacerbate brain injury and functional deficits. Although stem cell therapy is considered to be an alternative treatment for brain injuries, such as TBI and stroke, many obstacles remain. In particular, the time window for TBI treatment with either drugs or stem cells and their efficacy is still vague. Human placenta-derived mesenchymal stem cells (hpMSCs) have received extensive attention in stem cell therapy because they can be acquired in large numbers without ethical issues and because of their immune-modulating capacity and effectiveness in several diseases, such as Alzheimer’s disease and stroke. Here, we tested the feasibility of hpMSCs for TBI treatment with an animal model and attempted to identify appropriate time points for cell treatments. Double injections at 4 and 24 h post-injury significantly reduced the infarct size and suppressed astrocyte and microglial activation around the injury. With reduced damage, double-injected mice showed enhanced anti-inflammatory- and TNF-α receptor 2 (TNFR2)-associated survival signals and suppressed pro-inflammatory and oxidative responses. In addition, double-treated TBI mice displayed restored sensory motor functions and reduced neurotoxic Aβ42 plaque formation around the damaged areas. In this study, we showed the extended therapeutic potentials of hpMSCs and concluded that treatment within an appropriate time window is critical for TBI recovery.
Collapse
Affiliation(s)
- Chul Kim
- General Research Institute, CHA general Hospital, Seoul, South Korea
| | - Ji-Min Park
- Department of Biotechnology, College of Life Science, CHA University, Pangyo-ro 335 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, Seoul, South Korea.,General Research Institute, CHA general Hospital, Seoul, South Korea
| | - TaeHo Kong
- Department of Biotechnology, College of Life Science, CHA University, Pangyo-ro 335 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, Seoul, South Korea.,General Research Institute, CHA general Hospital, Seoul, South Korea
| | - Seungmin Lee
- General Research Institute, CHA general Hospital, Seoul, South Korea
| | - Ki-Weon Seo
- General Research Institute, CHA general Hospital, Seoul, South Korea.,SK Chemicals, Eco-Hub, 332 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13493, South Korea
| | - Yuri Choi
- Department of Biotechnology, College of Life Science, CHA University, Pangyo-ro 335 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, Seoul, South Korea
| | - Young Sook Song
- General Research Institute, CHA general Hospital, Seoul, South Korea
| | - Jisook Moon
- Department of Biotechnology, College of Life Science, CHA University, Pangyo-ro 335 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, Seoul, South Korea. .,General Research Institute, CHA general Hospital, Seoul, South Korea.
| |
Collapse
|
16
|
Salehi A, Zhang JH, Obenaus A. Response of the cerebral vasculature following traumatic brain injury. J Cereb Blood Flow Metab 2017; 37:2320-2339. [PMID: 28378621 PMCID: PMC5531360 DOI: 10.1177/0271678x17701460] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The critical role of the vasculature and its repair in neurological disease states is beginning to emerge particularly for stroke, dementia, epilepsy, Parkinson's disease, tumors and others. However, little attention has been focused on how the cerebral vasculature responds following traumatic brain injury (TBI). TBI often results in significant injury to the vasculature in the brain with subsequent cerebral hypoperfusion, ischemia, hypoxia, hemorrhage, blood-brain barrier disruption and edema. The sequalae that follow TBI result in neurological dysfunction across a host of physiological and psychological domains. Given the importance of restoring vascular function after injury, emerging research has focused on understanding the vascular response after TBI and the key cellular and molecular components of vascular repair. A more complete understanding of vascular repair mechanisms are needed and could lead to development of new vasculogenic therapies, not only for TBI but potentially vascular-related brain injuries. In this review, we delineate the vascular effects of TBI, its temporal response to injury and putative biomarkers for arterial and venous repair in TBI. We highlight several molecular pathways that may play a significant role in vascular repair after brain injury.
Collapse
Affiliation(s)
- Arjang Salehi
- 1 Cell, Molecular and Developmental Biology Program, University of California, Riverside, CA, USA.,2 Department of Pediatrics, Loma Linda University, Loma Linda, CA, USA
| | - John H Zhang
- 3 Department of Physiology and Pharmacology Loma Linda University School of Medicine, CA, USA.,4 Department of Anesthesiology Loma Linda University School of Medicine, CA, USA.,5 Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Andre Obenaus
- 1 Cell, Molecular and Developmental Biology Program, University of California, Riverside, CA, USA.,2 Department of Pediatrics, Loma Linda University, Loma Linda, CA, USA.,6 Department of Pediatrics, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
17
|
Bogoslovsky T, Bernstock JD, Bull G, Gouty S, Cox BM, Hallenbeck JM, Maric D. Development of a systems-based in situ multiplex biomarker screening approach for the assessment of immunopathology and neural tissue plasticity in male rats after traumatic brain injury. J Neurosci Res 2017; 96:487-500. [PMID: 28463430 DOI: 10.1002/jnr.24054] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 02/08/2017] [Accepted: 03/02/2017] [Indexed: 12/12/2022]
Abstract
Traumatic brain injuries (TBIs) pose a massive burden of disease and continue to be a leading cause of morbidity and mortality throughout the world. A major obstacle in developing effective treatments is the lack of comprehensive understanding of the underlying mechanisms that mediate tissue damage and recovery after TBI. As such, our work aims to highlight the development of a novel experimental platform capable of fully characterizing the underlying pathobiology that unfolds after TBI. This platform encompasses an empirically optimized multiplex immunohistochemistry staining and imaging system customized to screen for a myriad of biomarkers required to comprehensively evaluate the extent of neuroinflammation, neural tissue damage, and repair in response to TBI. Herein, we demonstrate that our multiplex biomarker screening platform is capable of evaluating changes in both the topographical location and functional states of resident and infiltrating cell types that play a role in neuropathology after controlled cortical impact injury to the brain in male Sprague-Dawley rats. Our results demonstrate that our multiplex biomarker screening platform lays the groundwork for the comprehensive characterization of changes that occur within the brain after TBI. Such work may ultimately lead to the understanding of the governing pathobiology of TBI, thereby fostering the development of novel therapeutic interventions tailored to produce optimal tissue protection, repair, and/or regeneration with minimal side effects, and may ultimately find utility in a wide variety of other neurological injuries, diseases, and disorders that share components of TBI pathobiology.
Collapse
Affiliation(s)
- Tanya Bogoslovsky
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences (CNRM/USUHS), Rockville, Maryland
| | - Joshua D Bernstock
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NINDS/NIH), Bethesda, Maryland.,Department of Clinical Neurosciences, Division of Stem Cell Neurobiology, Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Greg Bull
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences (CNRM/USUHS), Rockville, Maryland.,Department of Pharmacology, Uniformed Services University of the Health Sciences (USUHS), Bethesda, Maryland
| | - Shawn Gouty
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences (CNRM/USUHS), Rockville, Maryland.,Department of Pharmacology, Uniformed Services University of the Health Sciences (USUHS), Bethesda, Maryland
| | - Brian M Cox
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences (CNRM/USUHS), Rockville, Maryland.,Department of Pharmacology, Uniformed Services University of the Health Sciences (USUHS), Bethesda, Maryland
| | - John M Hallenbeck
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NINDS/NIH), Bethesda, Maryland
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NINDS/NIH), Bethesda, Maryland
| |
Collapse
|
18
|
Dutta D, Fauer C, Hickey K, Salifu M, Stabenfeldt SE. Tunable delayed controlled release profile from layered polymeric microparticles. J Mater Chem B 2017; 5:4487-4498. [PMID: 28652916 DOI: 10.1039/c7tb00138j] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Composite microparticles (MPs) with layered architecture, engineered from poly(L-lactic acid) (PLLA) and poly(D,L-lactic-co-glycolic acid) (PLGA), are promising devices for achieving the delayed release of proteins. Here, we build on a water-in-oil-in-oil-in-water emulsion method of fabricating layered MPs with an emphasis on modulating the delay period of the protein release profile. Particle hardening parameters (i.e. polymer precipitation rate and total hardening time) following water-in-oil-in-oil-in-water emulsions are known to affect MP structure such as the core/shell material and cargo localization. We demonstrate that layered MPs fabricated with two different solvent evaporation parameters not only alter polymer and protein distribution within the hardened MPs, but also affect their protein release profiles. Secondly, we hypothesize that ethanol (EtOH), a semi-polar solvent miscible in both the solvent (dichloromethane; DCM) and non-solvent aqueous phases, likely alters DCM and water flux from the dispersed oil phase. The results reveal that EtOH affects protein distribution within MPs, and may also influence MP structural properties such as porosity and polymer distribution. To our knowledge, we are the first to demonstrate EtOH as a means for modulating critical release parameters from protein-loaded, layered PLGA/PLLA MPs. Throughout all the groups in the study, we achieved differential delay periods (between 0 - 30 days after an initial burst release) and total protein release periods (~30 - >58 days) as a function of solvent evaporation parameters and EtOH content. The layered MPs proposed in the study potentially have wide-reaching applications in tissue engineering for delayed and sequential protein release.
Collapse
Affiliation(s)
- D Dutta
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - C Fauer
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - K Hickey
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - M Salifu
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - S E Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
19
|
Blood-brain barrier breakdown and neovascularization processes after stroke and traumatic brain injury. Curr Opin Neurol 2016; 28:556-64. [PMID: 26402408 DOI: 10.1097/wco.0000000000000248] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Angiogenesis or vascular reorganization plays a role in recovery after stroke and traumatic brain injury (TBI). In this review, we have focused on two major events that occur during stroke and TBI from a vascular perspective - what is the process and time course of blood-brain barrier (BBB) breakdown? and how does the surrounding vasculature recover and facilitate repair? RECENT FINDINGS Despite differences in the primary injury, the BBB changes overlap between stroke and TBI. Disruption of BBB involves a series of events: formation of caveolae, trans and paracellular disruption, tight junction breakdown and vascular disruption. Confounding factors that need careful assessment and standardization are the severity, duration and extent of the stroke and TBI that influences BBB disruption. Vascular repair proceeds through long-term neovascularization processes: angiogenesis, arteriogenesis and vasculogenesis. Enhancing each of these processes may impart beneficial effects in endogenous recovery. SUMMARY Our understanding of BBB breakdown acutely after the cerebrovascular injury has come a long way; however, we lack a clear understanding of the course of BBB disruption and BBB recovery and the evolution of individual cellular events associated with BBB change. Neovascularization responses have been widely studied in stroke for their role in functional recovery but the role of vascular reorganization after TBI in recovery is much less defined.
Collapse
|
20
|
Boccazzi M, Ceruti S. Where do you come from and what are you going to become, reactive astrocyte? Stem Cell Investig 2016; 3:15. [PMID: 27488400 DOI: 10.21037/sci.2016.05.02] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 04/27/2016] [Indexed: 11/06/2022]
Affiliation(s)
- Marta Boccazzi
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9 - 20133 Milan, Italy
| | - Stefania Ceruti
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9 - 20133 Milan, Italy
| |
Collapse
|
21
|
Dutta D, Fauer C, Mulleneux HL, Stabenfeldt SE. Tunable Controlled Release of Bioactive SDF-1α via Protein Specific Interactions within Fibrin/Nanoparticle Composites. J Mater Chem B 2015; 3:7963-7973. [PMID: 26660666 DOI: 10.1039/c5tb00935a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The chemokine, stromal cell-derived factor 1α (SDF-1α), is a key regulator of the endogenous neural progenitor/stem cell-mediated regenerative response after neural injury. Increased and sustained bioavailability of SDF-1α in the peri-injury region is hypothesized to modulate this endogenous repair response. Here, we describe poly(lactic-co-glycolic) acid (PLGA) nanoparticles capable of releasing bioactive SDF-1α in a sustained manner over 60days after a burst of 23%. Moreover, we report a biphasic cellular response to SDF-1α concentrations thus the large initial burst release in an in vivo setting may result in supratherapeutic concentrations of SDF-1α. Specific protein-protein interactions between SDF-1α and fibrin (as well as its monomer, fibrinogen) were exploited to control the magnitude of the burst release. Nanoparticles embedded in fibrin significantly reduced the amount of SDF-1α released after 72 hrs as a function of fibrin density. Therefore, the nanoparticle/fibrin composites represented a means to independently tune the magnitude of the burst phase release from the nanoparticles while perserving a bioactive depot of SDF-1α for release over 60days.
Collapse
Affiliation(s)
- D Dutta
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - C Fauer
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - H L Mulleneux
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - S E Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| |
Collapse
|