1
|
Ghasemi M, Mehranfard N. Neuroprotective actions of norepinephrine in neurological diseases. Pflugers Arch 2024; 476:1703-1725. [PMID: 39136758 DOI: 10.1007/s00424-024-02999-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/24/2024] [Accepted: 07/24/2024] [Indexed: 10/09/2024]
Abstract
Precise control of norepinephrine (NE) levels and NE-receptor interaction is crucial for proper function of the brain. Much evidence for this view comes from experimental studies that indicate an important role for NE in the pathophysiology and treatment of various conditions, including cognitive dysfunction, Alzheimer's disease, Parkinson's disease, multiple sclerosis, and sleep disorders. NE provides neuroprotection against several types of insults in multiple ways. It abrogates oxidative stress, attenuates neuroinflammatory responses in neurons and glial cells, reduces neuronal and glial cell activity, promotes autophagy, and ameliorates apoptotic responses to a variety of insults. It is beneficial for the treatment of neurodegenerative diseases because it improves the generation of neurotrophic factors, promotes neuronal survival, and plays an important role in the regulation of adult neurogenesis. This review aims to present the evidence supporting a principal role for NE in neuroprotection, and molecular mechanisms of neuroprotection.
Collapse
Affiliation(s)
- Maedeh Ghasemi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nasrin Mehranfard
- Nanokadeh Darooee Samen Private Joint Stock Company, Shafa Street, Urmia, 5715793731, Iran.
| |
Collapse
|
2
|
Wilson KL, Joseph NI, Onweller LA, Anderson AR, Darling NJ, David-Bercholz J, Segura T. SDF-1 Bound Heparin Nanoparticles Recruit Progenitor Cells for Their Differentiation and Promotion of Angiogenesis after Stroke. Adv Healthc Mater 2024; 13:e2302081. [PMID: 38009291 PMCID: PMC11128481 DOI: 10.1002/adhm.202302081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/22/2023] [Indexed: 11/28/2023]
Abstract
Angiogenesis after stroke is correlated with enhanced tissue repair and functional outcomes. The existing body of research in biomaterials for stroke focuses on hydrogels for the delivery of stem cells, growth factors, or small molecules or drugs. Despite the ability of hydrogels to enhance all these delivery methods, no material has significantly regrown vasculature within the translatable timeline of days to weeks after stroke. Here, two novel biomaterial formulations of granular hydrogels are developed for tissue regeneration after stroke: highly porous microgels (i.e., Cryo microgels) and microgels bound with heparin-norbornene nanoparticles with covalently bound SDF-1α. The combination of these materials results in perfused vessels throughout the stroke core in only 10 days, in addition to increased neural progenitor cell recruitment, maintenance, and increased neuronal differentiation.
Collapse
Affiliation(s)
- Katrina L. Wilson
- Department of Biomedical Engineering, Duke University, Durham NC 27708-0281, USA
| | - Neica I. Joseph
- Department of Biomedical Engineering, Duke University, Durham NC 27708-0281, USA
| | - Lauren A. Onweller
- Department of Biomedical Engineering, Duke University, Durham NC 27708-0281, USA
| | - Alexa R. Anderson
- Department of Biomedical Engineering, Duke University, Durham NC 27708-0281, USA
| | - Nicole J. Darling
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | | | - Tatiana Segura
- Department of Biomedical Engineering, Duke University, Durham NC 27708-0281, USA
- Department of Neurology, Duke University, Durham, NC, 27708-0281 USA
- Department of Dermatology, Duke University, Durham, NC, 27708-0281 USA
| |
Collapse
|
3
|
Santos CL, Weber FB, Belló-Klein A, Bobermin LD, Quincozes-Santos A. Glioprotective Effects of Sulforaphane in Hypothalamus: Focus on Aging Brain. Neurochem Res 2024; 49:2505-2518. [PMID: 38886329 DOI: 10.1007/s11064-024-04196-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 06/04/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
Sulforaphane is a natural compound with neuroprotective activity, but its effects on hypothalamus remain unknown. In line with this, astrocytes are critical cells to maintain brain homeostasis, and hypothalamic astrocytes are fundamental for sensing and responding to environmental changes involved in a variety of homeostatic functions. Changes in brain functionality, particularly associated with hypothalamic astrocytes, can contribute to age-related neurochemical alterations and, consequently, neurodegenerative diseases. Thus, here, we investigated the glioprotective effects of sulforaphane on hypothalamic astrocyte cultures and hypothalamic cell suspension obtained from aged Wistar rats (24 months old). Sulforaphane showed anti-inflammatory and antioxidant properties, as well as modulated the mRNA expression of astroglial markers, such as aldehyde dehydrogenase 1 family member L1, aquaporin 4, and vascular endothelial growth factor. In addition, it increased the expression and extracellular levels of trophic factors, such as glia-derived neurotrophic factor and nerve growth factor, as well as the release of brain-derived neurotrophic factor and the mRNA of TrkA, which is a receptor associated with trophic factors. Sulforaphane also modulated the expression of classical pathways associated with glioprotection, including nuclear factor erythroid-derived 2-like 2, heme oxygenase-1, nuclear factor kappa B p65 subunit, and AMP-activated protein kinase. Finally, a cell suspension with neurons and glial cells was used to confirm the predominant effect of sulforaphane in glial cells. In summary, this study indicated the anti-aging and glioprotective activities of sulforaphane in aged astrocytes.
Collapse
Affiliation(s)
- Camila Leite Santos
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fernanda Becker Weber
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Adriane Belló-Klein
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Larissa Daniele Bobermin
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - André Quincozes-Santos
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Laboratório de Neurotoxicidade e Glioproteção (LABGLIO), Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600 - Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
4
|
Loiola RA, Hachani J, Duban-Deweer S, Sevin E, Bugno P, Kowalska A, Rizzi E, Shimizu F, Kanda T, Mysiorek C, Mazurek M, Gosselet F. Secretome of brain microvascular endothelial cells promotes endothelial barrier tightness and protects against hypoxia-induced vascular leakage. Mol Med 2024; 30:132. [PMID: 39187765 PMCID: PMC11348522 DOI: 10.1186/s10020-024-00897-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024] Open
Abstract
Cell-based therapeutic strategies have been proposed as an alternative for brain and blood vessels repair after stroke, but their clinical application is hampered by potential adverse effects. We therefore tested the hypothesis that secretome of these cells might be used instead to still focus on cell-based therapeutic strategies. We therefore characterized the composition and the effect of the secretome of brain microvascular endothelial cells (BMECs) on primary in vitro human models of angiogenesis and vascular barrier. Two different secretome batches produced in high scale (scHSP) were analysed by mass spectrometry. Human primary CD34+-derived endothelial cells (CD34+-ECs) were used as well as in vitro models of EC monolayer (CMECs) and blood-brain barrier (BBB). Cells were also exposed to oxygen-glucose deprivation (OGD) conditions and treated with scHSP during reoxygenation. Protein yield and composition of scHSP batches showed good reproducibility. scHSP increased CD34+-EC proliferation, tubulogenesis, and migration. Proteomic analysis of scHSP revealed the presence of growth factors and proteins modulating cell metabolism and inflammatory pathways. scHSP improved the integrity of CMECs, and upregulated the expression of junctional proteins. Such effects were mediated through the activation of the interferon pathway and downregulation of Wnt signalling. Furthermore, OGD altered the permeability of both CMECs and BBB, while scHSP prevented the OGD-induced vascular leakage in both models. These effects were mediated through upregulation of junctional proteins and regulation of MAPK/VEGFR2. Finally, our results highlight the possibility of using secretome from BMECs as a therapeutic alternative to promote brain angiogenesis and to protect from ischemia-induced vascular leakage.
Collapse
Affiliation(s)
- Rodrigo Azevedo Loiola
- UR 2465, Laboratory of the Blood-Brain Barrier (LBHE), Sciences Faculty Jean Perrin, Artois University, 62300, Lens, France
| | - Johan Hachani
- UR 2465, Laboratory of the Blood-Brain Barrier (LBHE), Sciences Faculty Jean Perrin, Artois University, 62300, Lens, France
| | - Sophie Duban-Deweer
- UR 2465, Laboratory of the Blood-Brain Barrier (LBHE), Sciences Faculty Jean Perrin, Artois University, 62300, Lens, France
| | - Emmanuel Sevin
- UR 2465, Laboratory of the Blood-Brain Barrier (LBHE), Sciences Faculty Jean Perrin, Artois University, 62300, Lens, France
| | - Paulina Bugno
- Pure Biologics S.A., Duńska 11, 54-427, Wroclaw, Poland
| | | | - Eleonora Rizzi
- UR 2465, Laboratory of the Blood-Brain Barrier (LBHE), Sciences Faculty Jean Perrin, Artois University, 62300, Lens, France
| | - Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Caroline Mysiorek
- UR 2465, Laboratory of the Blood-Brain Barrier (LBHE), Sciences Faculty Jean Perrin, Artois University, 62300, Lens, France
| | | | - Fabien Gosselet
- UR 2465, Laboratory of the Blood-Brain Barrier (LBHE), Sciences Faculty Jean Perrin, Artois University, 62300, Lens, France.
| |
Collapse
|
5
|
Song XY, Fan CX, Atta-ur-Rahman FRS, Choudhary MI, Wang XP. Neuro-regeneration or Repair: Cell Therapy of Neurological Disorders as A Way Forward. Curr Neuropharmacol 2024; 22:2272-2283. [PMID: 38939990 PMCID: PMC11451317 DOI: 10.2174/1570159x22666240509092903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/28/2024] [Accepted: 05/02/2024] [Indexed: 06/29/2024] Open
Abstract
The human central nervous system (CNS) has a limited capacity for regeneration and repair, as many other organs do. Partly as a result, neurological diseases are the leading cause of medical burden globally. Most neurological disorders cannot be cured, and primary treatments focus on managing their symptoms and slowing down their progression. Cell therapy for neurological disorders offers several therapeutic potentials and provides hope for many patients. Here we provide a general overview of cell therapy in neurological disorders such as Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), Wilson's disease (WD), stroke and traumatic brain injury (TBI), involving many forms of stem cells, including embryonic stem cells and induced pluripotent stem cells. We also address the current concerns and perspectives for the future. Most studies for cell therapy in neurological diseases are in the pre-clinical stage, and there is still a great need for further research to translate neural replacement and regenerative therapies into clinical settings.
Collapse
Affiliation(s)
- Xiao-Yan Song
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Cun-Xiu Fan
- Department of Neurology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Atta-ur-Rahman FRS
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Muhammad Iqbal Choudhary
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Xiao-Ping Wang
- Department of Neurology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Bautista-Perez SM, Silva-Islas CA, Sandoval-Marquez OU, Toledo-Toledo J, Bello-Martínez JM, Barrera-Oviedo D, Maldonado PD. Antioxidant and Anti-Inflammatory Effects of Garlic in Ischemic Stroke: Proposal of a New Mechanism of Protection through Regulation of Neuroplasticity. Antioxidants (Basel) 2023; 12:2126. [PMID: 38136245 PMCID: PMC10740829 DOI: 10.3390/antiox12122126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/02/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Stroke represents one of the main causes of death and disability in the world; despite this, pharmacological therapies against stroke remain insufficient. Ischemic stroke is the leading etiology of stroke. Different molecular mechanisms, such as excitotoxicity, oxidative stress, and inflammation, participate in cell death and tissue damage. At a preclinical level, different garlic compounds have been evaluated against these mechanisms. Additionally, there is evidence supporting the participation of garlic compounds in other mechanisms that contribute to brain tissue recovery, such as neuroplasticity. After ischemia, neuroplasticity is activated to recover cognitive and motor function. Some garlic-derived compounds and preparations have shown the ability to promote neuroplasticity under physiological conditions and, more importantly, in cerebral damage models. This work describes damage/repair mechanisms and the importance of garlic as a source of antioxidant and anti-inflammatory agents against damage. Moreover, we examine the less-explored neurotrophic properties of garlic, culminating in proposals and observations based on our review of the available information. The aim of the present study is to propose that garlic compounds and preparations could contribute to the treatment of ischemic stroke through their neurotrophic effects.
Collapse
Affiliation(s)
- Sandra Monserrat Bautista-Perez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (S.M.B.-P.); (J.M.B.-M.); (D.B.-O.)
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (C.A.S.-I.); (O.U.S.-M.); (J.T.-T.)
| | - Carlos Alfredo Silva-Islas
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (C.A.S.-I.); (O.U.S.-M.); (J.T.-T.)
| | - Oscar Uriel Sandoval-Marquez
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (C.A.S.-I.); (O.U.S.-M.); (J.T.-T.)
| | - Jesús Toledo-Toledo
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (C.A.S.-I.); (O.U.S.-M.); (J.T.-T.)
- Servicio de Cirugía General, Hospital General de Zona #30, Instituto Mexicano del Seguro Social, Mexico City 08300, Mexico
| | - José Manuel Bello-Martínez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (S.M.B.-P.); (J.M.B.-M.); (D.B.-O.)
- Departamento Cirugía General, Hospital Central Militar, Mexico City 11600, Mexico
| | - Diana Barrera-Oviedo
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (S.M.B.-P.); (J.M.B.-M.); (D.B.-O.)
| | - Perla D. Maldonado
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (S.M.B.-P.); (J.M.B.-M.); (D.B.-O.)
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (C.A.S.-I.); (O.U.S.-M.); (J.T.-T.)
| |
Collapse
|
7
|
Wilson KL, Onweller LA, Joseph NI, David-Bercholz J, Darling NJ, Segura T. SDF-1 Bound Heparin Nanoparticles Recruit Progenitor Cells for Their Differentiation and Promotion of Angiogenesis After Stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.05.547800. [PMID: 37461490 PMCID: PMC10349963 DOI: 10.1101/2023.07.05.547800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Angiogenesis after stroke is correlated with enhanced tissue repair and functional outcomes. The existing body of research in biomaterials for stroke focuses on hydrogels for the delivery of stem cells, growth factors, or small molecules or drugs. Despite the ability of hydrogels to enhance all these delivery methods, no material has significantly regrown vasculature within the translatable timeline of days to weeks after stroke. Here we developed 2 novel biomaterials for tissue regeneration after stroke, a highly porous granular hydrogel termed Cryo microgels, and heparin-norbornene nanoparticles with covalently bound SDF-1α. The combination of these materials resulted in fully revascularized vessels throughout the stroke core in only 10 days, as well as increased neural progenitor cell migration and maintenance and increased neurons.
Collapse
|
8
|
Carmona-Mora P, Knepp B, Jickling GC, Zhan X, Hakoupian M, Hull H, Alomar N, Amini H, Sharp FR, Stamova B, Ander BP. Monocyte, neutrophil, and whole blood transcriptome dynamics following ischemic stroke. BMC Med 2023; 21:65. [PMID: 36803375 PMCID: PMC9942321 DOI: 10.1186/s12916-023-02766-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 12/21/2022] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND After ischemic stroke (IS), peripheral leukocytes infiltrate the damaged region and modulate the response to injury. Peripheral blood cells display distinctive gene expression signatures post-IS and these transcriptional programs reflect changes in immune responses to IS. Dissecting the temporal dynamics of gene expression after IS improves our understanding of immune and clotting responses at the molecular and cellular level that are involved in acute brain injury and may assist with time-targeted, cell-specific therapy. METHODS The transcriptomic profiles from peripheral monocytes, neutrophils, and whole blood from 38 ischemic stroke patients and 18 controls were analyzed with RNA-seq as a function of time and etiology after stroke. Differential expression analyses were performed at 0-24 h, 24-48 h, and >48 h following stroke. RESULTS Unique patterns of temporal gene expression and pathways were distinguished for monocytes, neutrophils, and whole blood with enrichment of interleukin signaling pathways for different time points and stroke etiologies. Compared to control subjects, gene expression was generally upregulated in neutrophils and generally downregulated in monocytes over all times for cardioembolic, large vessel, and small vessel strokes. Self-organizing maps identified gene clusters with similar trajectories of gene expression over time for different stroke causes and sample types. Weighted Gene Co-expression Network Analyses identified modules of co-expressed genes that significantly varied with time after stroke and included hub genes of immunoglobulin genes in whole blood. CONCLUSIONS Altogether, the identified genes and pathways are critical for understanding how the immune and clotting systems change over time after stroke. This study identifies potential time- and cell-specific biomarkers and treatment targets.
Collapse
Affiliation(s)
- Paulina Carmona-Mora
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA.
| | - Bodie Knepp
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| | - Glen C Jickling
- Division of Neurology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, 87 Avenue & 114 Street, Edmonton, AB, T6G 2J7, Canada
| | - Xinhua Zhan
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| | - Marisa Hakoupian
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| | - Heather Hull
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| | - Noor Alomar
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| | - Hajar Amini
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| | - Frank R Sharp
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| | - Boryana Stamova
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| | - Bradley P Ander
- Department of Neurology and M.I.N.D, Institute, M.I.N.D. Institute Bioscience Labs, School of Medicine, University of California at Davis, 2805 50th St, Room 2434, Sacramento, CA, 95817, USA
| |
Collapse
|
9
|
Goraltchouk A, Mankovskaya S, Kuznetsova T, Hladkova Z, Hollander JM, Luppino F, Seregin A. Comparative evaluation of rhFGF18 and rhGDF11 treatment in a transient ischemia stroke model. Restor Neurol Neurosci 2023; 41:257-270. [PMID: 38363623 DOI: 10.3233/rnn-231347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Background Pharmacological treatments for ischemic stroke remain limited to thrombolysis, which is associated with increased risk of potentially fatal hemorrhage. Treatments with Recombinant Human Fibroblast Growth Factor 18 (rhFGF18) and Growth and Differentiation Factor 11 (rhGDF11) appear promising based on different preclinical models. The goal of this study was to compare the effects of rhFGF18 and rhGDF11 directly on survival, behavioral deficits, and histological fingerprint of cerebral ischemia in the Wistar rat middle cerebral artery occlusion (MCAO) model of stroke. Methods Ischemia-reperfusion injury was induced using a 2-hour transient MCAO. Animals were administered rhFGF18 (infusion), rhGDF11 (multi-injection), or Phosphate Buffered Saline (PBS) vehicle control and followed for 42 days. Motor-Cognitive deficits were evaluated using the Morris Water Maze at Days 0 (pre-MCAO), 7, 21, and 42. Histopathological assessments were performed on Days 21 and 42. Results Day 7 post-ischemia water maze performance times increased 38.3%, 2.1%, and 23.1% for PBS, rhFGF18, and rhGDF11-treated groups, respectively. Fraction of neurons with abnormal morphology (chromatolysis, pyknotic nuclei, somal degeneration) decreased in all groups toward Day 42 and was lowest for rhFGF18. AChE-positive fiber density and activity increased over time in the rhFGF18 group, remained unchanged in the rhGDF11 treatment arm, and declined in the PBS control. Metabolic increases were greatest in rhGDF11 treated animals, with both rhFGF18 and rhGDF11 achieving improvements over PBS, as evidenced by increased succinate dehydrogenase and lactate dehydrogenase activity. Finally, rhFGF18 treatment exhibited a trend for reduced mortality relative to PBS (5.6%, 95% CI [27.3%, 0.1% ] vs. 22.2%, 95% CI [47.6%, 6.4% ]). Conclusions rhFGF18 treatment appears promising in improving survival and promoting motor-cognitive recovery following cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
| | | | | | - Zhanna Hladkova
- Institute of Physiology, National Academy of Sciences, Minsk, Belarus
| | - Judith M Hollander
- Remedium Bio, Inc., Needham, MA, USA
- Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | | | | |
Collapse
|
10
|
Bui TA, Jickling GC, Winship IR. Neutrophil dynamics and inflammaging in acute ischemic stroke: A transcriptomic review. Front Aging Neurosci 2022; 14:1041333. [PMID: 36620775 PMCID: PMC9813499 DOI: 10.3389/fnagi.2022.1041333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Stroke is among the leading causes of death and disability worldwide. Restoring blood flow through recanalization is currently the only acute treatment for cerebral ischemia. Unfortunately, many patients that achieve a complete recanalization fail to regain functional independence. Recent studies indicate that activation of peripheral immune cells, particularly neutrophils, may contribute to microcirculatory failure and futile recanalization. Stroke primarily affects the elderly population, and mortality after endovascular therapies is associated with advanced age. Previous analyses of differential gene expression across injury status and age identify ischemic stroke as a complex age-related disease. It also suggests robust interactions between stroke injury, aging, and inflammation on a cellular and molecular level. Understanding such interactions is crucial in developing effective protective treatments. The global stroke burden will continue to increase with a rapidly aging human population. Unfortunately, the mechanisms of age-dependent vulnerability are poorly defined. In this review, we will discuss how neutrophil-specific gene expression patterns may contribute to poor treatment responses in stroke patients. We will also discuss age-related transcriptional changes that may contribute to poor clinical outcomes and greater susceptibility to cerebrovascular diseases.
Collapse
Affiliation(s)
- Truong An Bui
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Glen C. Jickling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, Division of Neurology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ian R. Winship
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
11
|
Wang Z, Wang X, Liao Y, Chen G, Xu K. Immune response treated with bone marrow mesenchymal stromal cells after stroke. Front Neurol 2022; 13:991379. [PMID: 36203971 PMCID: PMC9530191 DOI: 10.3389/fneur.2022.991379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Stroke is a leading cause of death and long-term disability worldwide. Tissue plasminogen activator (tPA) is an effective treatment for ischemic stroke. However, only a small part of patients could benefit from it. Therefore, finding a new treatment is necessary. Bone marrow mesenchymal stromal cells (BMSCs) provide a novel strategy for stroke patients. Now, many patients take stem cells to treat stroke. However, the researches of the precise inflammatory mechanism of cell replacement treatment are still rare. In this review, we summarize the immune response of BMSCs treated to stroke and may provide a new perspective for stem cell therapy.
Collapse
Affiliation(s)
- Zili Wang
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Xudong Wang
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Yidong Liao
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Guangtang Chen
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Kaya Xu
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
- *Correspondence: Kaya Xu
| |
Collapse
|
12
|
Mousavi SM, Akbarpour B, Karimi-Haghighi S, Pandamooz S, Belém-Filho IJA, Masís-Calvo M, Salimi H, Lashanizadegan R, Pouramini A, Owjfard M, Hooshmandi E, Bayat M, Zafarmand SS, Dianatpour M, Salehi MS, Borhani-Haghighi A. Therapeutic potential of hair follicle-derived stem cell intranasal transplantation in a rat model of ischemic stroke. BMC Neurosci 2022; 23:47. [PMID: 35879657 PMCID: PMC9316709 DOI: 10.1186/s12868-022-00732-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 07/15/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Stem cell-based therapy has received considerable attention as a potential candidate in the treatment of ischemic stroke; however, employing an appropriate type of stem cells and an effective delivery route are still challenging. In the present study, we investigated the therapeutic effect of safe, noninvasive, and brain-targeted intranasal administration of hair follicle-derived stem cells (HFSCs) in a rat model of ischemic stroke. METHODS Stem cells were obtained from the adult rat hair follicles. In experiment 1, stroke was induced by 30 min middle cerebral artery occlusion (MCAO) and stem cells were intranasally transplanted immediately after ischemia. In experiment 2, stroke was induced by 120 min MCAO and stem cells were administered 24 h after cerebral ischemia. In all experimental groups, neurological performance, short-term spatial working memory and infarct volume were assessed. Moreover, relative expression of major trophic factors in the striatum and cortex was evaluated by the quantitative PCR technique. The end point of experiment 1 was day 3 and the end point of experiment 2 was day 15. RESULTS In both experiments, intranasal administration of HFSCs improved functional performance and decreased infarct volume compared to the MCAO rats. Furthermore, NeuN and VEGF expression were higher in the transplanted group and stem cell therapy partially prevented BDNF and neurotrophin-3 over-expression induced by cerebral ischemia. CONCLUSIONS These findings highlight the curative potential of HFSCs following intranasal transplantation in a rat model of ischemic stroke.
Collapse
Affiliation(s)
- Seyedeh Maryam Mousavi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bijan Akbarpour
- Department of Basic Sciences, Faculty of Veterinary Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran.
| | | | - Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Haniye Salimi
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ramin Lashanizadegan
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Pouramini
- Department of Basic Sciences, Faculty of Veterinary Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Maryam Owjfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Etrat Hooshmandi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahnaz Bayat
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mehdi Dianatpour
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | | |
Collapse
|
13
|
Øverberg LT, Lugg EF, Gaarder M, Langhammer B, Thommessen B, Rønning OM, Morland C. Plasma levels of BDNF and EGF are reduced in acute stroke patients. Heliyon 2022; 8:e09661. [PMID: 35756121 PMCID: PMC9218156 DOI: 10.1016/j.heliyon.2022.e09661] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/22/2022] [Accepted: 05/31/2022] [Indexed: 12/22/2022] Open
Abstract
Stroke affects almost 14 million people worldwide each year. It is the second leading cause of death and a major cause of acquired disability. The degree of initial impairment in cognitive and motor functions greatly affects the recovery, but idiosyncratic factors also contribute. These are largely unidentified, which contributes to making accurate prediction of recovery challenging. Release of soluble regulators of neurotoxicity, neuroprotection and repair are presumably essential. Here we measured plasma levels of known regulators of neuroprotection and repair in patients with mild acute ischemic stroke and compared them to the plasma levels in healthy age and gender matched controls. We found that the levels of BDNF and EGF were substantially lower in stroke patients than in healthy controls, while the levels of bFGF and irisin did not differ between the groups. The lower levels of growth factors highlight that during the acute phase of stroke, there is a mismatch between the need for neuroprotection and repair, and the brain's ability to induce these processes. Large individual differences in growth factor levels were seen among the stroke patients, but whether these can be used as predictors of long-term prognosis remains to be investigated.
Collapse
Affiliation(s)
- Linda Thøring Øverberg
- Department of Behavioral Sciences, Faculty of Health Sciences, OsloMet-Oslo Metropolitan University, Oslo, Norway.,Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, The Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Elise Fritsch Lugg
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, The Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Mona Gaarder
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, The Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Birgitta Langhammer
- Department of Physiotherapy, Faculty of Health Sciences, OsloMet-Oslo Metropolitan University, Oslo, Norway.,Research Department, Sunnaas Rehabilitation Hospital, Nesoddtangen, Norway
| | - Bente Thommessen
- Department of Neurology, Division of Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Ole Morten Rønning
- Department of Neurology, Division of Medicine, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Cecilie Morland
- Department of Behavioral Sciences, Faculty of Health Sciences, OsloMet-Oslo Metropolitan University, Oslo, Norway.,Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, The Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
14
|
Patel D, Wairkar S. Biotechnology-based therapeutics for management of cerebral stroke. Eur J Pharmacol 2021; 913:174638. [PMID: 34801531 DOI: 10.1016/j.ejphar.2021.174638] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023]
Abstract
Cerebral stroke, commonly caused due to hindrance in blood flow, is broadly classified into two categories-ischemic and haemorrhagic strokes. The onset of stroke triggers multiple mechanisms causing inflammation, generation of free radicals and protein damage leading to apoptosis of neuronal cells. The current therapies available for cerebral strokes involve use of complex surgical treatments and tissue plasminogen activator which increases the risk of internal bleeding, brain edema and cerebral damage, thereby restricting their use in clinical setting. The alarming need to develop safe, effective, target specific systems which, promote neuronal growth and reduce cerebral inflammation can be accomplished with use of biotechnological approaches. The article gives an insight to biotechnology-based advancements for tissue plasminogen activators, cell penetrating peptides, growth factors, ribonucleic acid systems and monoclonal antibodies for cerebral stroke. We also emphasis on challenges and future perspective of biotechnology-based therapeutics for better management of stroke.
Collapse
Affiliation(s)
- Dhrumi Patel
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India.
| |
Collapse
|
15
|
Chitu V, Biundo F, Stanley ER. Colony stimulating factors in the nervous system. Semin Immunol 2021; 54:101511. [PMID: 34743926 DOI: 10.1016/j.smim.2021.101511] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/23/2021] [Indexed: 01/02/2023]
Abstract
Although traditionally seen as regulators of hematopoiesis, colony-stimulating factors (CSFs) have emerged as important players in the nervous system, both in health and disease. This review summarizes the cellular sources, patterns of expression and physiological roles of the macrophage (CSF-1, IL-34), granulocyte-macrophage (GM-CSF) and granulocyte (G-CSF) colony stimulating factors within the nervous system, with a particular focus on their actions on microglia. CSF-1 and IL-34, via the CSF-1R, are required for the development, proliferation and maintenance of essentially all CNS microglia in a temporal and regional specific manner. In contrast, in steady state, GM-CSF and G-CSF are mainly involved in regulation of microglial function. The alterations in expression of these growth factors and their receptors, that have been reported in several neurological diseases, are described and the outcomes of their therapeutic targeting in mouse models and humans are discussed.
Collapse
Affiliation(s)
- Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Fabrizio Biundo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - E Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
16
|
Loiola RA, García-Gabilondo M, Grayston A, Bugno P, Kowalska A, Duban-Deweer S, Rizzi E, Hachani J, Sano Y, Shimizu F, Kanda T, Mysiorek C, Mazurek MP, Rosell A, Gosselet F. Secretome of endothelial progenitor cells from stroke patients promotes endothelial barrier tightness and protects against hypoxia-induced vascular leakage. Stem Cell Res Ther 2021; 12:552. [PMID: 34702368 PMCID: PMC8549346 DOI: 10.1186/s13287-021-02608-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/25/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Cell-based therapeutic strategies have been proposed as an alternative for brain repair after stroke, but their clinical application has been hampered by potential adverse effects in the long term. The present study was designed to test the effect of the secretome of endothelial progenitor cells (EPCs) from stroke patients (scCM) on in vitro human models of angiogenesis and vascular barrier. METHODS Two different scCM batches were analysed by mass spectrometry and a proteome profiler. Human primary CD34+-derived endothelial cells (CD34+-ECs) were used for designing angiogenesis studies (proliferation, migration, and tubulogenesis) or in vitro models of EC monolayer (confluent monolayer ECs-CMECs) and blood-brain barrier (BBB; brain-like ECs-BLECs). Cells were treated with scCM (5 μg/mL) or protein-free endothelial basal medium (scEBM-control). CMECs or BLECs were exposed (6 h) to oxygen-glucose deprivation (OGD) conditions (1% oxygen and glucose-free medium) or normoxia (control-5% oxygen, 1 g/L of glucose) and treated with scCM or scEBM during reoxygenation (24 h). RESULTS The analysis of different scCM batches showed a good reproducibility in terms of protein yield and composition. scCM increased CD34+-EC proliferation, tubulogenesis, and migration compared to the control (scEBM). The proteomic analysis of scCM revealed the presence of growth factors and molecules modulating cell metabolism and inflammatory pathways. Further, scCM decreased the permeability of CMECs and upregulated the expression of the junctional proteins such as occludin, VE-cadherin, and ZO-1. Such effects were possibly mediated through the activation of the interferon pathway and a moderate downregulation of Wnt signalling. Furthermore, OGD increased the permeability of both CMECs and BLECs, while scCM prevented the OGD-induced vascular leakage in both models. These effects were possibly mediated through the upregulation of junctional proteins and the regulation of MAPK/VEGFR2 activity. CONCLUSION Our results suggest that scCM promotes angiogenesis and the maturation of newly formed vessels while restoring the BBB function in ischemic conditions. In conclusion, our results highlight the possibility of using EPC-secretome as a therapeutic alternative to promote brain angiogenesis and protect from ischemia-induced vascular leakage.
Collapse
Affiliation(s)
| | - Miguel García-Gabilondo
- Neurovascular Research Laboratory, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, 08035, Barcelona, Catalonia, Spain
| | - Alba Grayston
- Neurovascular Research Laboratory, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, 08035, Barcelona, Catalonia, Spain
| | - Paulina Bugno
- Pure Biologics S.A., Duńska 11, 54-427, Wroclaw, Poland
| | | | - Sophie Duban-Deweer
- UR 2465, Blood-Brain Barrier Laboratory (LBHE), Univ. Artois, 62300, Lens, France
| | - Eleonora Rizzi
- UR 2465, Blood-Brain Barrier Laboratory (LBHE), Univ. Artois, 62300, Lens, France
| | - Johan Hachani
- UR 2465, Blood-Brain Barrier Laboratory (LBHE), Univ. Artois, 62300, Lens, France
| | - Yasuteru Sano
- Department of Neurology and Clinical Neuroscience, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Caroline Mysiorek
- UR 2465, Blood-Brain Barrier Laboratory (LBHE), Univ. Artois, 62300, Lens, France
| | | | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, 08035, Barcelona, Catalonia, Spain
| | - Fabien Gosselet
- UR 2465, Blood-Brain Barrier Laboratory (LBHE), Univ. Artois, 62300, Lens, France.
- Laboratory of the Blood-Brain Barrier, Sciences Faculty Jean Perrin, Artois University, Lens, France.
| |
Collapse
|
17
|
Zhang S, Shang D, Shi H, Teng W, Tian L. Function of Astrocytes in Neuroprotection and Repair after Ischemic Stroke. Eur Neurol 2021; 84:426-434. [PMID: 34455410 DOI: 10.1159/000517378] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/12/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Astrocytes are the most numerous cell types within the central nervous system, and many efforts have been put into determining the exact role of astrocytes in neuroprotection and repair after ischemic stroke. Although numerous studies have been done in recent years, there is still no thorough understanding of the exact function of astrocytes in the whole course of the stroke. SUMMARY According to the recent literature, there are many structures and factors that play important roles in the process of ischemic stroke, among which blood-brain barrier, various growth factors, gap junctions, AQP4, and glial scars have been studied most comprehensively, and all these factors are closely related to astrocytes. The role of astrocytes in ischemic stroke, therefore, can be analyzed more comprehensively. Key Message: The present review mainly summarized the current knowledge about astrocytes and their potential roles after ischemic stroke.
Collapse
Affiliation(s)
- Shufen Zhang
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, China,
| | - Deshu Shang
- Cell Biology Division, Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Han Shi
- The First Clinical College, China Medical University, Shenyang, China
| | - Weiyu Teng
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Li Tian
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
18
|
Asgharzade S, Talaei A, Farkhondeh T, Forouzanfar F. Combining Growth Factor and Stem Cell Therapy for Stroke Rehabilitation, A Review. Curr Drug Targets 2021; 21:781-791. [PMID: 31914912 DOI: 10.2174/1389450121666200107100747] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/28/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022]
Abstract
Stroke is a serious, life-threatening condition demanding vigorous search for new therapies. Recent research has focused on stem cell-based therapies as a viable choice following ischemic stroke, based on studies displaying that stem cells transplanted to the brain not only survive but also cause functional recovery. Growth factors defined as polypeptides that regulate the growth and differentiation of many cell types. Many studies have demonstrated that combined use of growth factors may increase results by the stimulation of endogenous neurogenesis, anti-inflammatory, neuroprotection properties, and enhancement of stem cell survival rates and so may be more effective than a single stem cell therapy. This paper reviews and discusses the most promising new stroke recovery research, including combination treatment.
Collapse
Affiliation(s)
- Samira Asgharzade
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Andisheh Talaei
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
19
|
Wang HK, Chen JS, Hsu CY, Su YT, Sung TC, Liang CL, Kwan AL, Wu CC. A Novel NGF Receptor Agonist B355252 Ameliorates Neuronal Loss and Inflammatory Responses in a Rat Model of Cerebral Ischemia. J Inflamm Res 2021; 14:2363-2376. [PMID: 34103967 PMCID: PMC8179829 DOI: 10.2147/jir.s303833] [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: 01/26/2021] [Accepted: 05/13/2021] [Indexed: 01/19/2023] Open
Abstract
Introduction Cerebral ischemia is a leading cause of disability and death worldwide. However, an effective therapeutic approach for the condition remains undiscovered. The previously proposed growth factor-based therapy has been inefficient due to its inability to pass through the blood–brain barrier. B355252, a newly developed small molecule, exhibited a potential neuroprotective effect in vivo. However, its exact efficacy in cerebral ischemia remains unclear. Methods We adopt an endothelin-1 stereotaxic intracranial injection to induced cerebral ischemia in rat. We further conducted 2,3,5-triphenyltetrazolium chloride (TTC) staining, immunofluorescent staining, enzyme-linked immunosorbent assay (ELISA), and behavioral tests to evaluate the efficacy of B355252 in neuroprotection, anti-inflammation, and behavioral outcome improvements. Results We identified that B355252 could protect ischemic neurons from neuronal loss by attenuating DNA damage, reducing ROS production and the LDH level, and preventing neuronal apoptosis. Moreover, inflammatory responses in astrocytic and microglial gliosis, as well as IL-1β and TNF-α levels, were ameliorated. Consequently, the behavioral outcomes of ischemic rats in neurologic responses and fore paw function recovery were improved. Discussion Overall, our study verified the in vivo therapeutic potential of B355252. The study findings further support its application in the development of a therapeutic approach for stroke.
Collapse
Affiliation(s)
- Hao-Kuang Wang
- Department of Neurosurgery, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan.,School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Jui-Sheng Chen
- Department of Neurosurgery, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Neurosurgery, E-Da Dachang Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Chien-Yu Hsu
- Department of Neurosurgery, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Yu-Ting Su
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Tzu-Ching Sung
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Cheng-Loong Liang
- Department of Neurosurgery, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Aij-Lie Kwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Cheng-Chun Wu
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| |
Collapse
|
20
|
Dordoe C, Chen K, Huang W, Chen J, Hu J, Wang X, Lin L. Roles of Fibroblast Growth Factors and Their Therapeutic Potential in Treatment of Ischemic Stroke. Front Pharmacol 2021; 12:671131. [PMID: 33967812 PMCID: PMC8102031 DOI: 10.3389/fphar.2021.671131] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 03/31/2021] [Indexed: 11/13/2022] Open
Abstract
Stroke is the leading cause of death worldwide, and its treatment remains a challenge. Complex pathological processes are involved in stroke, which causes a reduction in the supply of oxygen and energy to the brain that triggers subsequent cascade events, such as oxidative stress, inflammatory responses and apoptosis, resulting in brain injury. Stroke is a devastating disease for which there are few treatments, but physical rehabilitation can help improve stroke recovery. Although there are very few treatments for stroke patients, the discovery of fibroblast growth factors (FGFs) in mammals has led to the finding that FGFs can effectively treat stroke in animal models. As presented in this review, FGFs play essential roles by functioning as homeostatic factors and controlling cells and hormones involved in metabolism. They could be used as effective therapeutic agents for stroke. In this review, we will discuss the pharmacological actions of FGFs on multiple targets, including their ability to directly promote neuron survival, enhance angiogenesis, protect against blood-brain barrier (BBB) disruption, and regulate microglial modulation, in the treatment of ischemic stroke and their theoretical mechanisms and actions, as well as the therapeutic potential and limitations of FGFs for the clinical treatment of stroke.
Collapse
Affiliation(s)
- Confidence Dordoe
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Keyang Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Department of Neurology, The Second Affiliated Hospital and Yuying Children' Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenting Huang
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jun Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jian Hu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xue Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Li Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Research Units of Clinical Translation of Cell Growth Factors and Diseases Research, Chinese Academy of Medical Science, Beijing, China
| |
Collapse
|
21
|
Khan H, Singh A, Thapa K, Garg N, Grewal AK, Singh TG. Therapeutic modulation of the phosphatidylinositol 3-kinases (PI3K) pathway in cerebral ischemic injury. Brain Res 2021; 1761:147399. [PMID: 33662337 DOI: 10.1016/j.brainres.2021.147399] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 02/09/2021] [Accepted: 02/23/2021] [Indexed: 12/16/2022]
Abstract
The cerebral ischemic reperfusion injury may leads to morbidity and mortality in patients. phosphatidylinositol 3-kinase (PI3K) signaling pathway has been believed to work in association with its downstream targets, other receptors, and pathways that may offer antioxidant, anti-inflammatory, anti-apoptotic effects, neuroprotective role in neuronal excitotoxicity. This review elaborates the mechanistic interventions of the PI3K pathway in cerebral ischemic injury in context to nuclear factor erythroid 2-related factor 2 (Nrf2) regulation, Hypoxia-inducible factor 1 signaling (HIF-1), growth factors, Endothelial NOS (eNOS) proinflammatory cytokines, Erythropoietin (EPO), Phosphatase and tensin homologous protein of chromosome 10 gene (PTEN) signaling, NF-κB/Notch signaling, c-Jun N-terminal kinase (JNK) and Glycogen synthase kinase-3β (GSK-3β) signaling pathway. Evidences showing the activation of PI3K inhibits apoptotic pathway, which results in its neuroprotective effect in ischemic injury. Despite discussing the therapeutic role of the PI3K pathway in treating cerebral ischemic injury, the review also enlighten the selective modulation of PI3K pathway with activators and inhibitors which may provide promising results in clinical and preclinical settings.
Collapse
Affiliation(s)
- Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Anjali Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Komal Thapa
- Chitkara College of Pharmacy, Chitkara University, Punjab, India; School of Pharmacy, Chitkara University, Himachal Pradesh, India
| | - Nikhil Garg
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | | |
Collapse
|
22
|
Drug delivery platforms for neonatal brain injury. J Control Release 2021; 330:765-787. [PMID: 33417984 DOI: 10.1016/j.jconrel.2020.12.056] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 12/18/2022]
Abstract
Hypoxic-ischemic encephalopathy (HIE), initiated by the interruption of oxygenated blood supply to the brain, is a leading cause of death and lifelong disability in newborns. The pathogenesis of HIE involves a complex interplay of excitotoxicity, inflammation, and oxidative stress that results in acute to long term brain damage and functional impairments. Therapeutic hypothermia is the only approved treatment for HIE but has limited effectiveness for moderate to severe brain damage; thus, pharmacological intervention is explored as an adjunct therapy to hypothermia to further promote recovery. However, the limited bioavailability and the side-effects of systemic administration are factors that hinder the use of the candidate pharmacological agents. To overcome these barriers, therapeutic molecules may be packaged into nanoscale constructs to enable their delivery. Yet, the application of nanotechnology in infants is not well examined, and the neonatal brain presents unique challenges. Novel drug delivery platforms have the potential to magnify therapeutic effects in the damaged brain, mitigate side-effects associated with high systemic doses, and evade mechanisms that remove the drugs from circulation. Encouraging pre-clinical data demonstrates an attenuation of brain damage and increased structural and functional recovery. This review surveys the current progress in drug delivery for treating neonatal brain injury.
Collapse
|
23
|
Perrelli A, Fatehbasharzad P, Benedetti V, Ferraris C, Fontanella M, De Luca E, Moglianetti M, Battaglia L, Retta SF. Towards precision nanomedicine for cerebrovascular diseases with emphasis on Cerebral Cavernous Malformation (CCM). Expert Opin Drug Deliv 2021; 18:849-876. [PMID: 33406376 DOI: 10.1080/17425247.2021.1873273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Cerebrovascular diseases encompass various disorders of the brain vasculature, such as ischemic/hemorrhagic strokes, aneurysms, and vascular malformations, also affecting the central nervous system leading to a large variety of transient or permanent neurological disorders. They represent major causes of mortality and long-term disability worldwide, and some of them can be inherited, including Cerebral Cavernous Malformation (CCM), an autosomal dominant cerebrovascular disease linked to mutations in CCM1/KRIT1, CCM2, or CCM3/PDCD10 genes.Areas covered: Besides marked clinical and etiological heterogeneity, some commonalities are emerging among distinct cerebrovascular diseases, including key pathogenetic roles of oxidative stress and inflammation, which are increasingly recognized as major disease hallmarks and therapeutic targets. This review provides a comprehensive overview of the different clinical features and common pathogenetic determinants of cerebrovascular diseases, highlighting major challenges, including the pressing need for new diagnostic and therapeutic strategies, and focusing on emerging innovative features and promising benefits of nanomedicine strategies for early detection and targeted treatment of such diseases.Expert opinion: Specifically, we describe and discuss the multiple physico-chemical features and unique biological advantages of nanosystems, including nanodiagnostics, nanotherapeutics, and nanotheranostics, that may help improving diagnosis and treatment of cerebrovascular diseases and neurological comorbidities, with an emphasis on CCM disease.
Collapse
Affiliation(s)
- Andrea Perrelli
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy.,CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy
| | - Parisa Fatehbasharzad
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy.,CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy
| | - Valerio Benedetti
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy.,CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy
| | - Chiara Ferraris
- Department of Drug Science and Technology, University of Torino, Torino, Italy.,Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, University of Torino, Torino, Italy
| | - Marco Fontanella
- CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy.,Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Elisa De Luca
- Nanobiointeractions & Nanodiagnostics, Center for Biomolecular Nanotechnologies, Arnesano, Lecce, Italy.,Institute for Microelectronics and Microsystems (IMM), CNR, Lecce, Italy
| | - Mauro Moglianetti
- Nanobiointeractions & Nanodiagnostics, Center for Biomolecular Nanotechnologies, Arnesano, Lecce, Italy.,Istituto Italiano Di Tecnologia, Nanobiointeractions & Nanodiagnostics, Genova, Italy
| | - Luigi Battaglia
- Department of Drug Science and Technology, University of Torino, Torino, Italy.,Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, University of Torino, Torino, Italy
| | - Saverio Francesco Retta
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy.,CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy
| |
Collapse
|
24
|
CXCR7, CXCR4, and Their Ligand Expression Profile in Traumatic Brain Injury. World Neurosurg 2020; 147:e16-e24. [PMID: 33189916 DOI: 10.1016/j.wneu.2020.11.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/05/2020] [Accepted: 11/05/2020] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Traumatic brain injury (TBI) is a health problem worldwide, and therapeutic strategies to enhance brain tissue repair to lessen neurologic sequels are imperative. We aimed to analyze the impact of the inflammatory process in TBI through CXCR4 and CXCR7 chemokine receptors and their ligands' CXCL11 and CXCL12 expression profile in search for potential new druggable targets. METHODS Twelve pericontusional tissues from severe TBI patients submitted to surgical treatment, and 20 control brain tissues from normal autopsy were analyzed for expression profile by real-time quantitative-polymerase chain reaction. CXCR7 and CXCR4 protein expressions were analyzed by immunohistochemistry. The findings were correlated with the clinical evolution. RESULTS Increased gene expression of both receptors and their ligands was observed in TBI compared with controls, presenting high sensitivity and specificity to differentiate TBI from normal control (area under the curve ranging from 0.85 to 0.98, P < 0.001). In particular, CXCR7 expression highly correlated with CXCR4 and both ligands' expressions in TBI. Higher immunoreactions for CXCR7 and CXCR4 were identified in neurons and endothelial cells of TBI samples compared with controls. The patients presenting upregulated chemokine expression levels showed a trend toward favorable clinical evolution at up to 6 months of follow-up. CONCLUSIONS The neuroprotective trend of CXCR4, CXCR7, CXCL11, and CXCL12 in TBI observed in this initial analysis warrants further studies with more patients, analyzing the involved signaling pathways for the development of new therapeutic strategies for TBI.
Collapse
|
25
|
Woods D, Jiang Q, Chu XP. Monoclonal antibody as an emerging therapy for acute ischemic stroke. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2020; 12:95-106. [PMID: 32934765 PMCID: PMC7486556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 08/20/2020] [Indexed: 06/11/2023]
Abstract
Acute ischemic stroke (AIS) is the 5th leading cause of death and the leading cause of neurological disability in the United States. The oxygen and glucose deprivation associated with AIS not only leads to neuronal cell death, but also increases the inflammatory response, therefore decreasing the functional outcome of the brain. The only pharmacological intervention approved by the US Federal Food and Drug Administration for treatment of AIS is tissue plasminogen activator (t-PA), however, such treatment can only be given within 4.5 hours of the onset of stroke-like symptoms. This narrow time-range limits its therapeutic application. Administrating t-PA outside of the therapeutic window may induce detrimental rather than beneficial effects to stroke patients. In order to reduce the infarct volume of an AIS while increasing the time period for treatment, new treatments are essential. Emerging monoclonal antibody (mAb) therapies reveal great potential by targeting signaling pathways activated after an AIS. With successful application of mAb in the treatment of cancer, other therapeutic uses for mAb are currently being evaluated. In this review, we will focus on recent advances on AIS therapy by using mAb that targets the signaling cascades and endogenous molecules such as inflammation, growth factors, acid-sensing ion channels, and N-methyl-D-aspartate receptors. Therefore, developing specific mAb to target the signaling pathways of ischemic brain injury will benefit patients being treated for an AIS.
Collapse
Affiliation(s)
- Demi Woods
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City Kansas City, MO 64108, USA
| | - Qian Jiang
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City Kansas City, MO 64108, USA
| | - Xiang-Ping Chu
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City Kansas City, MO 64108, USA
| |
Collapse
|
26
|
Alam AB, Wu A, Power MC, West NA, Alonso A. Associations of serum uric acid with incident dementia and cognitive decline in the ARIC-NCS cohort. J Neurol Sci 2020; 414:116866. [PMID: 32387846 PMCID: PMC7293945 DOI: 10.1016/j.jns.2020.116866] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/16/2020] [Accepted: 04/24/2020] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Elevated serum uric acid (SUA) is associated with cardiovascular risk factors, which often contribute to dementia and dementia-like morbidity, yet several cross-sectional studies have shown protective associations with cognition, which would be consistent with other work showing benefits of elevated SUA through its antioxidant properties. METHODS We studied 11,169 participants free of dementia and cardiovascular disease from the Atherosclerosis Risk in Communities (ARIC) cohort. SUA was measured in blood samples collected in 1990-92, baseline for this study (age range 47-70 years). Incident dementia was ascertained based on clinical assessments in 2011-13 and 2016-17, surveillance based on dementia screeners conducted over telephone interviews, hospitalization discharge codes, and death certificates. Cognitive function was assessed up to four times between 1990 and 92 and 2016-17. We estimated the association of SUA, categorized into quartiles, with incidence of dementia using Cox regression models adjusting for potential confounders. The association between cognitive decline and SUA was assessed using generalized estimating equations. RESULTS Over a median follow-up period of 24.1 years, 2005 cases of dementia were identified. High baseline SUA was associated with incident dementia (HR, 1.29; 95% CI, 1.12, 1.47) when adjusted for sociodemographic variables. However, after further adjustment including cardiovascular risk factors, this relationship disappeared (HR, 1.03; 95% CI, 0.88, 1.21). Elevated baseline SUA was associated with faster cognitive decline even after further adjustment (25-year global z-score difference, -0.149; 95% CI, -0.246, -0.052). CONCLUSION Higher levels of mid-life SUA were associated with faster cognitive decline, but not necessarily with higher risk of dementia.
Collapse
Affiliation(s)
- Aniqa B Alam
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| | - Aozhou Wu
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Melinda C Power
- Department of Epidemiology and Biostatistics, Milken School of Public Health, George Washington University, Washington D.C., USA
| | - Nancy A West
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| |
Collapse
|
27
|
Barui AK, Jhelum P, Nethi SK, Das T, Bhattacharya D, B V, Karri S, Chakravarty S, Patra CR. Potential Therapeutic Application of Zinc Oxide Nanoflowers in the Cerebral Ischemia Rat Model through Neuritogenic and Neuroprotective Properties. Bioconjug Chem 2020; 31:895-906. [PMID: 32050064 DOI: 10.1021/acs.bioconjchem.0c00030] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Neuritogenesis, a complex process of the sprouting of neurites, plays a vital role in the structural and functional restoration of cerebral ischemia-injured neuronal tissue. Practically, there is no effective long-term treatment strategy for cerebral ischemia in clinical practice to date due to several limitations of conventional therapies, facilitating the urgency to develop new alternative therapeutic approaches. Herein, for the first time we report that pro-angiogenic nanomaterials, zinc oxide nanoflowers (ZONF), exhibit neuritogenic activity by elevating mRNA expression of different neurotrophins, following PI3K/Akt-MAPK/ERK signaling pathways. Further, ZONF administration to global cerebral ischemia-induced Fischer rats shows improved neurobehavior and enhanced synaptic plasticity of neurons via upregulation of Neurabin-2 and NT-3, revealing their neuroprotective activity. Altogether, this study offers the basis for exploitation of angio-neural cross talk of other pro-angiogenic nano/biomaterials for future advancement of alternative treatment strategies for cerebral ischemia, where neuritogenesis and neural repair are highly critical.
Collapse
Affiliation(s)
- Ayan Kumar Barui
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Priya Jhelum
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
| | - Susheel Kumar Nethi
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Tapatee Das
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Dwaipayan Bhattacharya
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
| | - Vinothkumar B
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
| | - Shailaja Karri
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
| | - Sumana Chakravarty
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Chitta Ranjan Patra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| |
Collapse
|
28
|
Dhir N, Medhi B, Prakash A, Goyal MK, Modi M, Mohindra S. Pre-clinical to Clinical Translational Failures and Current Status of Clinical Trials in Stroke Therapy: A Brief Review. Curr Neuropharmacol 2020; 18:596-612. [PMID: 31934841 PMCID: PMC7457423 DOI: 10.2174/1570159x18666200114160844] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/31/2019] [Accepted: 12/28/2019] [Indexed: 12/16/2022] Open
Abstract
In stroke (cerebral ischemia), despite continuous efforts both at the experimental and clinical level, the only approved pharmacological treatment has been restricted to tissue plasminogen activator (tPA). Stroke is the leading cause of functional disability and mortality throughout worldwide. Its pathophysiology starts with energy pump failure, followed by complex signaling cascade that ultimately ends in neuronal cell death. Ischemic cascade involves excessive glutamate release followed by raised intracellular sodium and calcium influx along with free radicals' generation, activation of inflammatory cytokines, NO synthases, lipases, endonucleases and other apoptotic pathways leading to cell edema and death. At the pre-clinical stage, several agents have been tried and proven as an effective neuroprotectant in animal models of ischemia. However, these agents failed to show convincing results in terms of efficacy and safety when the trials were conducted in humans following stroke. This article highlights the various agents which have been tried in the past but failed to translate into stroke therapy along with key points that are responsible for the lagging of experimental success to translational failure in stroke treatment.
Collapse
Affiliation(s)
| | - Bikash Medhi
- Address correspondence to this author at the Department of Pharmacology, Research Block B, 4th Floor, Room no 4043, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India; E-mail:
| | | | | | | | | |
Collapse
|
29
|
Regenhardt RW, Takase H, Lo EH, Lin DJ. Translating concepts of neural repair after stroke: Structural and functional targets for recovery. Restor Neurol Neurosci 2020; 38:67-92. [PMID: 31929129 PMCID: PMC7442117 DOI: 10.3233/rnn-190978] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stroke is among the most common causes of adult disability worldwide, and its disease burden is shifting towards that of a long-term condition. Therefore, the development of approaches to enhance recovery and augment neural repair after stroke will be critical. Recovery after stroke involves complex interrelated systems of neural repair. There are changes in both structure (at the molecular, cellular, and tissue levels) and function (in terms of excitability, cortical maps, and networks) that occur spontaneously within the brain. Several approaches to augment neural repair through enhancing these changes are under study. These include identifying novel drug targets, implementing rehabilitation strategies, and developing new neurotechnologies. Each of these approaches has its own array of different proposed mechanisms. Current investigation has emphasized both cellular and circuit-based targets in both gray and white matter, including axon sprouting, dendritic branching, neurogenesis, axon preservation, remyelination, blood brain barrier integrity, blockade of extracellular inhibitory signals, alteration of excitability, and promotion of new brain cortical maps and networks. Herein, we review for clinicians recovery after stroke, basic elements of spontaneous neural repair, and ongoing work to augment neural repair. Future study requires alignment of basic, translational, and clinical research. The field continues to grow while becoming more clearly defined. As thrombolysis changed stroke care in the 1990 s and thrombectomy in the 2010 s, the augmentation of neural repair and recovery after stroke may revolutionize care for these patients in the coming decade.
Collapse
Affiliation(s)
- Robert W Regenhardt
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| | - Hajime Takase
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| | - Eng H Lo
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| | - David J Lin
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
- Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| |
Collapse
|
30
|
Yasuhara T, Kawauchi S, Kin K, Morimoto J, Kameda M, Sasaki T, Bonsack B, Kingsbury C, Tajiri N, Borlongan CV, Date I. Cell therapy for central nervous system disorders: Current obstacles to progress. CNS Neurosci Ther 2019; 26:595-602. [PMID: 31622035 PMCID: PMC7248543 DOI: 10.1111/cns.13247] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/24/2019] [Accepted: 09/29/2019] [Indexed: 12/13/2022] Open
Abstract
Cell therapy for disorders of the central nervous system has progressed to a new level of clinical application. Various clinical studies are underway for Parkinson's disease, stroke, traumatic brain injury, and various other neurological diseases. Recent biotechnological developments in cell therapy have taken advantage of the technology of induced pluripotent stem (iPS) cells. The advent of iPS cells has provided a robust stem cell donor source for neurorestoration via transplantation. Additionally, iPS cells have served as a platform for the discovery of therapeutics drugs, allowing breakthroughs in our understanding of the pathology and treatment of neurological diseases. Despite these recent advances in iPS, adult tissue‐derived mesenchymal stem cells remain the widely used donor for cell transplantation. Mesenchymal stem cells are easily isolated and amplified toward the cells' unique trophic factor‐secretion property. In this review article, the milestone achievements of cell therapy for central nervous system disorders, with equal consideration on the present translational obstacles for clinic application, are described.
Collapse
Affiliation(s)
- Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Satoshi Kawauchi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Kyohei Kin
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Jun Morimoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Masahiro Kameda
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Tatsuya Sasaki
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Brooke Bonsack
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Chase Kingsbury
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Naoki Tajiri
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences and Medical School, Aichi, Japan
| | - Cesario V Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| |
Collapse
|
31
|
Farokhi-Sisakht F, Farhoudi M, Sadigh-Eteghad S, Mahmoudi J, Mohaddes G. Cognitive Rehabilitation Improves Ischemic Stroke-Induced Cognitive Impairment: Role of Growth Factors. J Stroke Cerebrovasc Dis 2019; 28:104299. [DOI: 10.1016/j.jstrokecerebrovasdis.2019.07.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/24/2019] [Accepted: 07/13/2019] [Indexed: 12/20/2022] Open
|
32
|
Zhang Y, Ma L, Su Y, Su L, Lan X, Wu D, Han S, Li J, Kvederis L, Corey S, Borlongan CV, Ji X. Hypoxia conditioning enhances neuroprotective effects of aged human bone marrow mesenchymal stem cell-derived conditioned medium against cerebral ischemia in vitro. Brain Res 2019; 1725:146432. [PMID: 31491422 DOI: 10.1016/j.brainres.2019.146432] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/27/2019] [Accepted: 09/01/2019] [Indexed: 12/24/2022]
Abstract
Therapeutic transplantation of autologous bone marrow mesenchymal stem cells (BMSCs) holds great promise for ischemic stroke, yet the efficacy is negatively impacted by aging. Here, we examined whether hypoxia conditioning could enhance aged human BMSCs-induced neuroprotection via secretome action. Primary cultured mouse neurons were exposed to oxygen glucose deprivation (OGD) to mimic ischemic stroke in vitro, then randomized into a hypoxia conditioned aged human BMSCs-conditioned medium (BMSC-hypoCM) versus normoxia conditioned (BMSC-norCM). After 22 h of reperfusion, cell viability was significantly increased in neurons treated with BMSC-hypoCM rather than BMSC-norCM. ELISA revealed that hypoxia conditioning enhanced vascular endothelial growth factor (VEGF) release into BMSC-derived CM. Blocking the VEGF receptor negated BMSC-hypoCM-induced protection for neurons against OGD insult. Altogether, our data indicates that hypoxia conditioning improves aged human BMSCs' therapeutic efficacy for neurons with ischemic challenge, in part via promoting secretion of VEGF.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Neurobiology, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing 100053, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Longhui Ma
- Department of Neurobiology, Capital Medical University, Beijing 100069, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Yuwen Su
- Department of Neurobiology, Capital Medical University, Beijing 100069, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Li Su
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xiaoxi Lan
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Di Wu
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing 100053, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Song Han
- Department of Neurobiology, Capital Medical University, Beijing 100069, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Junfa Li
- Department of Neurobiology, Capital Medical University, Beijing 100069, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Lauren Kvederis
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Sydney Corey
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Xunming Ji
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing 100053, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China; Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
33
|
Ceci M, Mariano V, Romano N. Zebrafish as a translational regeneration model to study the activation of neural stem cells and role of their environment. Rev Neurosci 2019; 30:45-66. [PMID: 30067512 DOI: 10.1515/revneuro-2018-0020] [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: 03/04/2018] [Accepted: 04/27/2018] [Indexed: 02/07/2023]
Abstract
The review is an overview of the current knowledge of neuronal regeneration properties in mammals and fish. The ability to regenerate the damaged parts of the nervous tissue has been demonstrated in all vertebrates. Notably, fish and amphibians have the highest capacity for neurogenesis, whereas reptiles and birds are able to only regenerate specific regions of the brain, while mammals have reduced capacity for neurogenesis. Zebrafish (Danio rerio) is a promising model of study because lesions in the brain or complete cross-section of the spinal cord are followed by an effective neuro-regeneration that successfully restores the motor function. In the brain and the spinal cord of zebrafish, stem cell activity is always able to re-activate the molecular programs required for central nervous system regeneration. In mammals, traumatic brain injuries are followed by reduced neurogenesis and poor axonal regeneration, often insufficient to functionally restore the nervous tissue, while spinal injuries are not repaired at all. The environment that surrounds the stem cell niche constituted by connective tissue and stimulating factors, including pro-inflammation molecules, seems to be a determinant in triggering stem cell proliferation and/or the trans-differentiation of connective elements (mainly fibroblasts). Investigating and comparing the neuronal regeneration in zebrafish and mammals may lead to a better understanding of the mechanisms behind neurogenesis, and the failure of the regenerative response in mammals, first of all, the role of inflammation, considered the main inhibitor of the neuronal regeneration.
Collapse
Affiliation(s)
- Marcello Ceci
- Department of Ecological and Biological Sciences, University of Tuscia, largo dell'Università, I-01100 Viterbo, Italy
| | - Vittoria Mariano
- Department of Fundamental Neurosciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Nicla Romano
- Department of Ecological and Biological Sciences, University of Tuscia, largo dell'Università, I-01100 Viterbo, Italy
| |
Collapse
|
34
|
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
|
35
|
Dief AE, Hassan PS, Hartmut O, Jirikowski GF. Neuronal and glial regeneration after focal cerebral ischemia in rat, an immunohistochemical and electron microscopical study. ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2018.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Abeer E. Dief
- Department of Physiology, University of Alexandria, Egypt
| | | | | | | |
Collapse
|
36
|
Central inhibition of granulocyte-macrophage colony-stimulating factor is analgesic in experimental neuropathic pain. Pain 2019; 159:550-559. [PMID: 29351125 PMCID: PMC5828377 DOI: 10.1097/j.pain.0000000000001130] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Supplemental Digital Content is Available in the Text. GM-CSF is a proinflammatory cytokine that plays a role in central pain pathways through the modulation of spinal glial cells. With less than 50% of patients responding to the current standard of care and poor efficacy and selectivity of current treatments, neuropathic pain continues to be an area of considerable unmet medical need. Biological therapeutics such as monoclonal antibodies (mAbs) provide better intrinsic selectivity; however, delivery to the central nervous system (CNS) remains a challenge. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is well described in inflammation-induced pain, and early-phase clinical trials evaluating its antagonism have exemplified its importance as a peripheral pain target. Here, we investigate the role of this cytokine in a murine model of traumatic nerve injury and show that deletion of the GM-CSF receptor or treatment with an antagonizing mAb alleviates pain. We also demonstrate enhanced analgesic efficacy using an engineered construct that has greater capacity to penetrate the CNS. Despite observing GM-CSF receptor expression in microglia and astrocytes, the gliosis response in the dorsal horn was not altered in nerve injured knockout mice compared with wild-type littermate controls as evaluated by ionized calcium binding adapter molecule 1 (Iba1) and glial fibrillary acidic protein, respectively. Functional analysis of glial cells revealed that pretreatment with GM-CSF potentiated lipopolysaccharide-induced release of proinflammatory cytokines. In summary, our data indicate that GM-CSF is a proinflammatory cytokine that contributes to nociceptive signalling through driving spinal glial cell secretion of proinflammatory mediators. In addition, we report a successful approach to accessing CNS pain targets, providing promise for central compartment delivery of analgesics.
Collapse
|
37
|
Goenka L, Uppugunduri Satyanarayana CR, S SK, George M. Neuroprotective agents in Acute Ischemic Stroke-A Reality Check. Biomed Pharmacother 2018; 109:2539-2547. [PMID: 30551514 DOI: 10.1016/j.biopha.2018.11.041] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 10/22/2018] [Accepted: 11/10/2018] [Indexed: 11/29/2022] Open
Affiliation(s)
- Luxitaa Goenka
- Department of Clinical Pharmacology, SRM Medical College Hospital & Research Centre, Kattankulathur, Chennai, Tamil Nadu, 603203 India
| | - Chakradhara Rao Uppugunduri Satyanarayana
- Platform of Pediatric Onco-Hematology (CANSEARCH Laboratory), Department of Pediatrics, University of Geneva, Bâtiment Tulipe, Avenue De La Roseraie, 641205 Geneva, Switzerland
| | - Suresh Kumar S
- Department of Pharmacology, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, Ras Al Khaymah, United Arab Emirates
| | - Melvin George
- Department of Clinical Pharmacology, SRM Medical College Hospital & Research Centre, Kattankulathur, Chennai, Tamil Nadu, 603203 India.
| |
Collapse
|
38
|
Uric Acid Protects against Focal Cerebral Ischemia/Reperfusion-Induced Oxidative Stress via Activating Nrf2 and Regulating Neurotrophic Factor Expression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6069150. [PMID: 30581534 PMCID: PMC6276484 DOI: 10.1155/2018/6069150] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/05/2018] [Accepted: 10/16/2018] [Indexed: 12/15/2022]
Abstract
The aim of this study was to investigate whether uric acid (UA) might exert neuroprotection via activating the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway and regulating neurotrophic factors in the cerebral cortices after transient focal cerebral ischemia/reperfusion (FCI/R) in rats. UA was intravenously injected through the tail vein (16 mg/kg) 30 min after the onset of reperfusion in rats subjected to middle cerebral artery occlusion for 2 h. Neurological deficit score was performed to analyze neurological function at 24 h after reperfusion. Terminal deoxynucleotidyl transferase-mediated dNTP nick end labeling (TUNEL) staining and hematoxylin and eosin (HE) staining were used to detect histological injury of the cerebral cortex. Malondialdehyde (MDA), the carbonyl groups, and 8-hydroxyl-2′-deoxyguanosine (8-OHdG) levels were employed to evaluate oxidative stress. Nrf2 and its downstream antioxidant protein, heme oxygenase- (HO-) 1,were detected by western blot. Nrf2 DNA-binding activity was observed using an ELISA-based measurement. Expressions of BDNF and NGF were analyzed by immunohistochemistry. Our results showed that UA treatment significantly suppressed FCI/R-induced oxidative stress, accompanied by attenuating neuronal damage, which subsequently decreased the infarct volume and neurological deficit. Further, the treatment of UA activated Nrf2 signaling pathway and upregulated BDNF and NGF expression levels. Interestingly, the aforementioned effects of UA were markedly inhibited by administration of brusatol, an inhibitor of Nrf2. Taken together, the antioxidant and neuroprotective effects afforded by UA treatment involved the modulation of Nrf2-mediated oxidative stress and regulation of BDNF and NGF expression levels. Thus, UA treatment could be of interest to prevent FCI/R injury.
Collapse
|
39
|
Khelif Y, Toutain J, Quittet MS, Chantepie S, Laffray X, Valable S, Divoux D, Sineriz F, Pascolo-Rebouillat E, Papy-Garcia D, Barritault D, Touzani O, Bernaudin M. A heparan sulfate-based matrix therapy reduces brain damage and enhances functional recovery following stroke. Am J Cancer Res 2018; 8:5814-5827. [PMID: 30613264 PMCID: PMC6299437 DOI: 10.7150/thno.28252] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/12/2018] [Indexed: 12/16/2022] Open
Abstract
Alteration of the extracellular matrix (ECM) is one of the major events in the pathogenesis of brain lesions following ischemic stroke. Heparan sulfate mimetics (HSm) are synthetic pharmacologically active polysaccharides that promote ECM remodeling and tissue regeneration in various types of lesions. HSm bind to growth factors, protect them from enzymatic degradation and increase their bioavailability, which promotes tissue repair. As the ECM is altered during stroke and HSm have been shown to restore the ECM, we investigated the potential of HSm4131 (also named RGTA-4131®) to protect brain tissue and promote regeneration and plasticity after a stroke. Methods: Ischemic stroke was induced in rats using transient (1 h) intraluminal middle cerebral artery occlusion (MCAo). Animals were assigned to the treatment (HSm4131; 0.1, 0.5, 1.5, or 5 mg/kg) or vehicle control (saline) groups at different times (1, 2.5 or 6 h) after MCAo. Brain damage was assessed by MRI for the acute (2 days) and chronic (14 days) phases post-occlusion. Functional deficits were evaluated with a battery of sensorimotor behavioral tests. HSm4131-99mTc biodistribution in the ischemic brain was analyzed between 5 min and 3 h following middle cerebral artery reperfusion. Heparan sulfate distribution and cellular reactions, including angiogenesis and neurogenesis, were evaluated by immunohistochemistry, and growth factor gene expression (VEGF-A, Ang-2) was quantified by RT-PCR. Results: HSm4131, administered intravenously after stroke induction, located and remained in the ischemic hemisphere. HSm4131 conferred long-lasting neuroprotection, and significantly reduced functional deficits with no alteration of physiological parameters. It also restored the ECM, and increased brain plasticity processes, i.e., angiogenesis and neurogenesis, in the affected brain hemisphere. Conclusion: HSm represent a promising ECM-based therapeutic strategy to protect and repair the brain after a stroke and favor functional recovery.
Collapse
|
40
|
Dames C, Winek K, Beckers Y, Engel O, Meisel A, Meisel C. Immunomodulatory treatment with systemic GM-CSF augments pulmonary immune responses and improves neurological outcome after experimental stroke. J Neuroimmunol 2018; 321:144-149. [DOI: 10.1016/j.jneuroim.2018.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/26/2018] [Accepted: 03/08/2018] [Indexed: 11/29/2022]
|
41
|
Zhang Y, Zhang H, Lin S, Chen X, Yao Y, Mao X, Shao B, Zhuge Q, Jin K. SDF-1/CXCR7 Chemokine Signaling is Induced in the Peri-Infarct Regions in Patients with Ischemic Stroke. Aging Dis 2018; 9:287-295. [PMID: 29896417 PMCID: PMC5963349 DOI: 10.14336/ad.2017.1112] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 11/12/2017] [Indexed: 12/11/2022] Open
Abstract
Stromal-derived factor-1 (SDF-1, also known as CXCL12) and its receptors CXCR4 and CXCR7 play important roles in brain repair after ischemic stroke, as SDF-1/ CXCR4/CXCR7 chemokine signaling is critical for recruiting stem cells to sites of ischemic injury. Upregulation of SDF-1/CXCR4/CXCR7 chemokine signaling in the ischemic regions has been well-documented in the animal models of ischemic stroke, but not in human ischemic brain. Here, we found that protein expression of SDF-1 and CXCR7, but not CXCR4, were significantly increased in the cortical peri-infarct regions (penumbra) after ischemic stroke in human, compared with adjacent normal tissues and control subjects. Double-label fluorescence immunohistochemistry shows that SDF-1 and CXCR4 proteins were expressed in neuronal cells and astrocytes in the normal brain tissue and peri-infarct regions. CXCR7 protein was also observed in neuronal cells and astrocytes in the normal cortical regions, but predominantly in astrocytes in the penumbra of ischemic brain. Our data suggest that ischemic stroke in human leads to an increase in the expression of SDF-1 and CXCR7, but not CXCR4, in the peri-infarct cerebral cortex. Our findings suggest that chemokine SFD-1 is expressed not only in animal models of stroke, but also in the human brain after an ischemic injury. In addition, unlike animals, CXCR7 may be the primary receptor of SDF-1 in human stroke brain.
Collapse
Affiliation(s)
- Yu Zhang
- 1Department of Neurosurgery, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Hongxia Zhang
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, Texas 76107, USA
| | - Siyang Lin
- 3Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xudong Chen
- 3Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yu Yao
- 4Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - XiaoOu Mao
- 5Buck Institute for Age Research, Novato, California 94945, USA
| | - Bei Shao
- 3Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Qichuan Zhuge
- 1Department of Neurosurgery, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.,3Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Kunlin Jin
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, Texas 76107, USA.,3Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| |
Collapse
|
42
|
Zhao LR, Willing A. Enhancing endogenous capacity to repair a stroke-damaged brain: An evolving field for stroke research. Prog Neurobiol 2018; 163-164:5-26. [PMID: 29476785 PMCID: PMC6075953 DOI: 10.1016/j.pneurobio.2018.01.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 01/11/2018] [Accepted: 01/30/2018] [Indexed: 02/07/2023]
Abstract
Stroke represents a severe medical condition that causes stroke survivors to suffer from long-term and even lifelong disability. Over the past several decades, a vast majority of stroke research targets neuroprotection in the acute phase, while little work has been done to enhance stroke recovery at the later stage. Through reviewing current understanding of brain plasticity, stroke pathology, and emerging preclinical and clinical restorative approaches, this review aims to provide new insights to advance the research field for stroke recovery. Lifelong brain plasticity offers the long-lasting possibility to repair a stroke-damaged brain. Stroke impairs the structural and functional integrity of entire brain networks; the restorative approaches containing multi-components have great potential to maximize stroke recovery by rebuilding and normalizing the stroke-disrupted entire brain networks and brain functioning. The restorative window for stroke recovery is much longer than previously thought. The optimal time for brain repair appears to be at later stage of stroke rather than the earlier stage. It is expected that these new insights will advance our understanding of stroke recovery and assist in developing the next generation of restorative approaches for enhancing brain repair after stroke.
Collapse
Affiliation(s)
- Li-Ru Zhao
- Department of Neurosurgery, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA.
| | - Alison Willing
- Center for Excellence in Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, 33612, USA.
| |
Collapse
|
43
|
Promoting neuroregeneration after perinatal arterial ischemic stroke: neurotrophic factors and mesenchymal stem cells. Pediatr Res 2018; 83:372-384. [PMID: 28949952 DOI: 10.1038/pr.2017.243] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/19/2017] [Indexed: 01/02/2023]
Abstract
Newborns suffering from perinatal arterial ischemic stroke (PAIS) are at risk of neurodevelopmental problems. Current treatment options for PAIS are limited and mainly focus on supportive care, as presentation of PAIS is beyond the time window of current treatment strategies. Therefore, recent focus has shifted to interventions that stimulate regeneration of damaged brain tissue. From animal models, it is known that the brain increases its neurogenic capability after ischemic injury, by promoting neural cell proliferation and differentiation. However, neurogenesis is not maintained at the long term, which consequently impedes full repair leading to adverse consequences later in life. Boosting neuroregeneration of the newborn brain using treatment with neurotrophic factors and/or mesenchymal stem cells (MSCs) may be promising novel therapeutic strategies to improve neurological prospects and quality of life of infants with PAIS. This review focuses on effectiveness of neurotrophic growth factors, including erythropoietin, brain-derived neurotrophic factor, vascular endothelial growth factor, glial-derived neurotrophic factor, and MSC therapy, in both experimental neonatal stroke studies and first clinical trials for neonatal ischemic brain injury.
Collapse
|
44
|
Cramer SC. Treatments to Promote Neural Repair after Stroke. J Stroke 2018; 20:57-70. [PMID: 29402069 PMCID: PMC5836581 DOI: 10.5853/jos.2017.02796] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 01/16/2018] [Accepted: 01/18/2018] [Indexed: 12/12/2022] Open
Abstract
Stroke remains a major cause of human disability worldwide. In parallel with advances in acute stroke interventions, new therapies are under development that target restorative processes. Such therapies have a treatment time window measured in days, weeks, or longer and so have the advantage that they may be accessible by a majority of patients. Several categories of restorative therapy have been studied and are reviewed herein, including drugs, growth factors, monoclonal antibodies, activity-related therapies including telerehabilitation, and a host of devices such as those related to brain stimulation or robotics. Many patients with stroke do not receive acute stroke therapies or receive them and do not derive benefit, often surviving for years thereafter. Therapies based on neural repair hold the promise of providing additional treatment options to a majority of patients with stroke.
Collapse
Affiliation(s)
- Steven C. Cramer
- Departments of Neurology, Anatomy & Neurobiology and Physical Medicine & Rehabilitation, University of California, Irvine, CA, USA
| |
Collapse
|
45
|
Leviton A, Allred EN, Yamamoto H, Fichorova RN, Kuban K, O'Shea TM, Dammann O. Antecedents and correlates of blood concentrations of neurotrophic growth factors in very preterm newborns. Cytokine 2017; 94:21-28. [PMID: 28396037 PMCID: PMC5464409 DOI: 10.1016/j.cyto.2017.03.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 01/18/2017] [Accepted: 03/31/2017] [Indexed: 11/16/2022]
Abstract
AIM To identify the antecedents and very early correlates of low concentrations of neurotrophic growth factors in the blood of extremely preterm newborns during the first postnatal month. METHODS Using an immunobead assay, we measured the concentrations of neurotrophin 4 (NT4), brain-derived neurotrophic factor (BDNF), and basic fibroblast growth factor (bFGF) in blood spots collected on postnatal days 1 (N=1062), 7 (N=1087), 14 (N=989), 21 (N=940) and 28 (N=880) from infants born before the 28th week of gestation. We then sought the correlates of measurements in the top and bottom quartiles for gestational age and day the specimen was collected. RESULTS The concentrations of 2 neurotrophic proteins, NT4 and BDNF, were low among children delivered for medical (maternal or fetal) indications, and among those who were growth restricted. Children who had top quartile concentrations of NT4, BDNF, and bFGF tended to have elevated concentrations of inflammation-related proteins that day. This pattern persisted for much of the first postnatal month. CONCLUSIONS Delivery for medical indications and fetal growth restriction are associated with a relative paucity of NT4 and BDNF concentrations during the first 24 h after very preterm birth. Elevated blood concentrations of NT4, BDNF, and bFGF tended to co-occur with indicators of systemic inflammation on the same day.
Collapse
Affiliation(s)
- Alan Leviton
- Boston Children's Hospital, and Harvard Medical School, Boston, MA, United States.
| | - Elizabeth N Allred
- Boston Children's Hospital, and Harvard Medical School, Boston, MA, United States
| | | | - Raina N Fichorova
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Karl Kuban
- Boston Medical Center and Boston University, Boston, MA, United States
| | | | - Olaf Dammann
- Tufts University School of Medicine, Boston, MA, United States; Hannover Medical School, Hannover, Germany
| |
Collapse
|
46
|
Khoshnam SE, Winlow W, Farzaneh M. The Interplay of MicroRNAs in the Inflammatory Mechanisms Following Ischemic Stroke. J Neuropathol Exp Neurol 2017; 76:548-561. [DOI: 10.1093/jnen/nlx036] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
47
|
Shen LH, Chen J, Shen HC, Ye M, Liu XF, Ding WS, Sheng YF, Ding XS. Possible Mechanism of Therapeutic Effect of 3-Methyl-1-phenyl-2-pyrazolin-5-one and Bone Marrow Stromal Cells Combination Treatment in Rat Ischemic Stroke Model. Chin Med J (Engl) 2017; 129:1471-6. [PMID: 27270545 PMCID: PMC4910373 DOI: 10.4103/0366-6999.183418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background: The functional improvement following bone marrow stromal cells (BMSCs) transplantation after stroke is directly related to the number of engrafted cells and neurogenesis in the injured brain. Here, we tried to evaluate whether 3-methyl-1-phenyl-2-pyrazolin-5-one (MCI-186), a free radical scavenger, might influence BMSCs migration to ischemic brain, which could promote neurogenesis and thereby enhance treatment effects after stroke. Methods: Rat transient middle cerebral artery occlusion (MCAO) model was established. Two separate MCAO groups were administered with either MCI-186 or phosphate-buffered saline (PBS) solution to evaluate the expression of stromal cell-derived factor-1 (SDF-1) in ischemic brain, and compared to that in sham group (n = 5/group/time point[at 1, 3, and 7 days after operation]). The content of chemokine receptor-4 (CXCR4, a main receptor of SDF-1) at 7 days after operation was also observed on cultured BMSCs. Another four MCAO groups were intravenously administered with either PBS, MCI-186, BMSCs (2 × 106), or a combination of MCI-186 and BMSCs (n = 10/group). 5-bromo-2-deoxyuridine (BrdU) and Nestin double-immunofluorescence staining was performed to identify the engrafted BMSCs and neuronal differentiation. Adhesive-removal test and foot-fault evaluation were used to test the neurological outcome. Results: MCI-186 upregulated the expression of SDF-1 in ischemic brain and CXCR4 content in BMSCs was enhanced after hypoxic stimulation. When MCAO rats were treated with either MCI-186, BMSCs, or a combination of MCI-186 and BMSCs, the neurologic function was obviously recovered as compared to PBS control group (P < 0.01 or 0.05, respectively). Combination therapy represented a further restoration, increased the number of BMSCs and Nestin+ cells in ischemic brain as compared with BMSCs monotherapy (P < 0.01). The number of engrafted-BMSCs was correlated with the density of neuronal cells in ischemic brain (r = 0.72, P < 0.01) and the improvement of foot-fault (r = 0.70, P < 0.01). Conclusion: MCI-186 might promote BMSCs migration to the ischemic brain, amplify the neurogenesis, and improve the effects of cell therapy.
Collapse
Affiliation(s)
- Li-Hua Shen
- Department of Neurology, The Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Jin Chen
- Department of Neurology, The Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Hua-Chao Shen
- The BenQ Neurological Institute of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Min Ye
- The BenQ Neurological Institute of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiao-Fei Liu
- Department of Neurology, The Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Wen-Sen Ding
- Department of Neurology, The Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Ya-Feng Sheng
- Department of Neurology, The Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Xin-Sheng Ding
- The BenQ Neurological Institute of Nanjing Medical University, Nanjing, Jiangsu 210029; Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
48
|
Cook DJ, Nguyen C, Chun HN, L Llorente I, Chiu AS, Machnicki M, Zarembinski TI, Carmichael ST. Hydrogel-delivered brain-derived neurotrophic factor promotes tissue repair and recovery after stroke. J Cereb Blood Flow Metab 2017; 37:1030-1045. [PMID: 27174996 PMCID: PMC5363479 DOI: 10.1177/0271678x16649964] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/20/2015] [Accepted: 03/20/2016] [Indexed: 11/15/2022]
Abstract
Stroke is the leading cause of adult disability. Systemic delivery of candidate neural repair therapies is limited by the blood-brain barrier and off-target effects. We tested a bioengineering approach for local depot release of BDNF from the infarct cavity for neural repair in chronic periods after stroke. The brain release levels of a hyaluronic acid hydrogel + BDNF were tested in several stroke models in mouse (strains C57Bl/6, DBA) and non-human primate ( Macaca fascicularis) and tracked with MRI. The behavioral recovery effects of hydrogel + BDNF and the effects on tissue repair outcomes were determined. Hydrogel-delivered BDNF diffuses from the stroke cavity into peri-infarct tissue over 3 weeks in two mouse stroke models, compared with 1 week for direct BDNF injection. Hydrogel delivery of BDNF promotes recovery of motor function. Mapping of motor system connections indicates that hydrogel-BDNF induces axonal sprouting within existing cortical and cortico-striatal systems. Pharmacogenetic studies show that hydrogel-BDNF induces the initial migration of immature neurons into the peri-infarct cortex and their long-term survival. In chronic stroke in the non-human primate, hydrogel-released BDNF can be detected up to 2 cm from the infarct, a distance relevant to human functional recovery in stroke. The hydrogel can be tracked by MRI in mouse and primate.
Collapse
Affiliation(s)
- Douglas J Cook
- Department of Surgery, Division of Neurosurgery, Kingston General Hospital, Kingston, Canada
| | - Cynthia Nguyen
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Hyun N Chun
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Irene L Llorente
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Abraham S Chiu
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Michal Machnicki
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | | | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| |
Collapse
|
49
|
Nguyen H, Aum D, Mashkouri S, Rao G, Vega Gonzales-Portillo JD, Reyes S, Borlongan CV. Growth factor therapy sequesters inflammation in affording neuroprotection in cerebrovascular diseases. Expert Rev Neurother 2016; 16:915-26. [PMID: 27152762 DOI: 10.1080/14737175.2016.1184086] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION In recent years, accumulating evidence has demonstrated the key role of inflammation in the progression of cerebrovascular diseases. Inflammation can persist over prolonged period of time after the initial insult providing a wider therapeutic window. Despite the acute endogenous upregulation of many growth factors after the injury, it is not sufficient to protect against inflammation and to regenerate the brain. Therapeutic approaches targeting both dampening inflammation and enhancing growth factors are likely to provide beneficial outcomes in cerebrovascular disease. AREAS COVERED In this mini review, we discuss major growth factors and their beneficial properties to combat the inflammation in cerebrovascular diseases. Emerging biotechnologies which facilitate the therapeutic effects of growth factors are also presented in an effort to provide insights into the future combination therapies incorporating both central and peripheral abrogation of inflammation. Expert commentary: Many studies discussed in this review have demonstrated the therapeutic effects of growth factors in treating cerebrovascular diseases. It is unlikely that one growth factor can be used to treat these complex diseases. Combination of growth factors and anti-inflammatory modulators may clinically improve outcomes for patients. In particular, transplantation of stem cells may be able to achieve both goals of modulating inflammation and upregulating growth factors. Large preclinical studies and multiple laboratory collaborations are needed to advance these findings from bench to bedside.
Collapse
Affiliation(s)
- Hung Nguyen
- a Department of Neurosurgery and Brain Repair , University of South Florida Morsani College of Medicine , Tampa , FL , USA
| | - David Aum
- a Department of Neurosurgery and Brain Repair , University of South Florida Morsani College of Medicine , Tampa , FL , USA
| | - Sherwin Mashkouri
- a Department of Neurosurgery and Brain Repair , University of South Florida Morsani College of Medicine , Tampa , FL , USA
| | - Gautam Rao
- a Department of Neurosurgery and Brain Repair , University of South Florida Morsani College of Medicine , Tampa , FL , USA
| | | | - Stephanny Reyes
- a Department of Neurosurgery and Brain Repair , University of South Florida Morsani College of Medicine , Tampa , FL , USA
| | - Cesario V Borlongan
- a Department of Neurosurgery and Brain Repair , University of South Florida Morsani College of Medicine , Tampa , FL , USA
| |
Collapse
|
50
|
|