1
|
Detante O, Legris L, Moisan A, Rome C. Cell Therapy and Functional Recovery of Stroke. Neuroscience 2024; 550:79-88. [PMID: 38013148 DOI: 10.1016/j.neuroscience.2023.11.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 11/29/2023]
Abstract
Stroke is the most common cause of disability. Brain repair mechanisms are often insufficient to allow a full recovery. Stroke damage involve all brain cell type and extracellular matrix which represent the crucial "glio-neurovascular niche" useful for brain plasticity. Regenerative medicine including cell therapies hold great promise to decrease post-stroke disability of many patients, by promoting both neuroprotection and neural repair through direct effects on brain lesion and/or systemic effects such as immunomodulation. Mechanisms of action vary according to each grafted cell type: "peripheral" stem cells, such as mesenchymal stem cells (MSC), can provide paracrine trophic support, and neural stem/progenitor cells (NSC) or neurons can act as direct cells' replacements. Optimal time window, route, and doses are still debated, and may depend on the chosen medicinal product and its expected mechanism such as neuroprotection, delayed brain repair, systemic effects, or graft survival and integration in host network. MSC, mononuclear cells (MNC), umbilical cord stem cells and NSC are the most investigated. Innovative approaches are implemented concerning combinatorial approaches with growth factors and biomaterials such as injectable hydrogels which could protect a cell graft and/or deliver drugs into the post-stroke cavity at chronic stages. Through main publications of the last two decades, we provide in this review concepts and suggestions to improve future translational researches and larger clinical trials of cell therapy in stroke.
Collapse
Affiliation(s)
- Olivier Detante
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institute Neurosciences, 38000 Grenoble, France; Stroke Unit, Neurology, CHU Grenoble Alpes, CS10217, 38043 Grenoble, France; Axe Neurosciences Cliniques - Innovative Brain Therapies, CHU Grenoble Alpes, 38000 Grenoble, France.
| | - Loic Legris
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institute Neurosciences, 38000 Grenoble, France; Stroke Unit, Neurology, CHU Grenoble Alpes, CS10217, 38043 Grenoble, France; Axe Neurosciences Cliniques - Innovative Brain Therapies, CHU Grenoble Alpes, 38000 Grenoble, France.
| | - Anaick Moisan
- Axe Neurosciences Cliniques - Innovative Brain Therapies, CHU Grenoble Alpes, 38000 Grenoble, France; Cell Therapy and Engineering Unit, EFS Rhône Alpes, 464 route de Lancey, 38330 Saint Ismier, France.
| | - Claire Rome
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institute Neurosciences, 38000 Grenoble, France; Stroke Unit, Neurology, CHU Grenoble Alpes, CS10217, 38043 Grenoble, France; Axe Neurosciences Cliniques - Innovative Brain Therapies, CHU Grenoble Alpes, 38000 Grenoble, France.
| |
Collapse
|
2
|
Sitruk-Ware R, Sussman H, Brinton R, Schumacher M, Singer P, Kumar N, De Nicola AF, El-Etr M, Guennoun R, V Borlongan C. Nestorone (segesterone acetate) effects on neuroregeneration. Front Neuroendocrinol 2024; 73:101136. [PMID: 38670433 DOI: 10.1016/j.yfrne.2024.101136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/08/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024]
Abstract
Nestorone® (segesterone acetate) is a progestin with a chemical structure closely related to progesterone with high affinity and selectivity for the progesterone receptor without significant interaction with other steroid receptors. It has been developed for female and male contraception and is FDA-approved in a first long-acting contraceptive vaginal system for female contraception. Its safety has been extensively demonstrated in both preclinical and clinical studies for contraceptive indications. Nestorone was found to display neuroprotective and neuroregenerative activity in animal models of various central nervous system diseases, including multiple sclerosis, stroke, and amyotrophic lateral sclerosis. Reviewed herein are neuroprotective and myelin- regenerating properties of Nestorone in various animal models and its translational potential as a therapeutic agent for debilitating neurological diseases for which limited therapeutic options are available (Table 1).
Collapse
Affiliation(s)
| | | | - Roberta Brinton
- Center for Innovation in Brain Science, Tucson, AZ, United States
| | | | | | | | | | - Martine El-Etr
- U1195 Inserm and University Paris-Saclay Le Kremlin Bicêtre, France
| | - Rachida Guennoun
- U1195 Inserm and University Paris-Saclay Le Kremlin Bicêtre, France
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
3
|
Karimi-Haghighi S, Pandamooz S, Jurek B, Fattahi S, Safari A, Azarpira N, Dianatpour M, Hooshmandi E, Bayat M, Owjfard M, Zafarmand SS, Mostaghel M, Mousavi SM, Jashire Nezhad N, Eraghi V, Fadakar N, Rahimi Jaberi A, Garcia-Esperon C, Spratt N, Levi C, Salehi MS, Borhani-Haghighi A. From Hair to the Brain: The Short-Term Therapeutic Potential of Human Hair Follicle-Derived Stem Cells and Their Conditioned Medium in a Rat Model of Stroke. Mol Neurobiol 2023; 60:2587-2601. [PMID: 36694047 DOI: 10.1007/s12035-023-03223-z] [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: 05/11/2022] [Accepted: 01/04/2023] [Indexed: 01/26/2023]
Abstract
The short-term therapeutic impacts of stem cells and their derivatives were frequently reported in preclinical investigations of ischemic stroke (IS); however, several drawbacks including accessibility, abundancy, and ethical concerns limited their clinical application. We describe here for the first time the therapeutic potential of human hair follicle-derived stem cells (hHFSCs) and their conditioned medium (CM) in a rat model of IS. Furthermore, we hypothesized that a combination of cell therapy with repeated CM administration might enhance the restorative efficiency of this approach compared to each treatment alone. Middle cerebral artery occlusion was performed for 30 min to induce IS. Immediately after reperfusion, hHFSCs were transplanted through the intra-arterial route and/or hHFSC-CM administered intranasally. The neurological outcomes, short-term spatial working memory, and infarct size were evaluated. Furthermore, relative expression of seven target genes in three categories of neuronal markers, synaptic markers, and angiogenic markers was assessed. The hHFSCs and hHFSC-CM treatments improved neurological impairments and reduced infarct size in the IS rats. Moreover, molecular data elucidated that IS was accompanied by attenuation in the expression of neuronal and synaptic markers in the evaluated brain regions and the interventions rescued these expression changes. Although there was no considerable difference between hHFSCs and hHFSC-CM treatments in the improvement of neurological function and decrement of infarct size, combination therapy was more effective to reduce infarction and elevation of target gene expression especially in the hippocampus. These findings highlight the curative potential of hHFSCs and their CM in a rat model of IS.
Collapse
Affiliation(s)
| | - Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Benjamin Jurek
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany
| | | | - Anahid Safari
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Dianatpour
- Stem Cells Technology 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
| | - Maryam Owjfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mandana Mostaghel
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Nahid Jashire Nezhad
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vida Eraghi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nima Fadakar
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Rahimi Jaberi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Carlos Garcia-Esperon
- Department of Neurology, John Hunter Hospital, Newcastle, Australia
- Hunter Medical Research Institute and the University of Newcastle, Newcastle, Australia
| | - Neil Spratt
- Department of Neurology, John Hunter Hospital, Newcastle, Australia
- Hunter Medical Research Institute and the University of Newcastle, Newcastle, Australia
| | - Christopher Levi
- Department of Neurology, John Hunter Hospital, Newcastle, Australia
- Hunter Medical Research Institute and the University of Newcastle, Newcastle, Australia
| | - Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Afshin Borhani-Haghighi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Neurology, John Hunter Hospital, Newcastle, Australia.
- Hunter Medical Research Institute and the University of Newcastle, Newcastle, Australia.
| |
Collapse
|
4
|
Berlet R, Galang Cabantan DA, Gonzales-Portillo D, Borlongan CV. Enriched Environment and Exercise Enhance Stem Cell Therapy for Stroke, Parkinson’s Disease, and Huntington’s Disease. Front Cell Dev Biol 2022; 10:798826. [PMID: 35309929 PMCID: PMC8927702 DOI: 10.3389/fcell.2022.798826] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/01/2022] [Indexed: 12/12/2022] Open
Abstract
Stem cells, specifically embryonic stem cells (ESCs), mesenchymal stem cells (MSCs), induced pluripotent stem cells (IPSCs), and neural progenitor stem cells (NSCs), are a possible treatment for stroke, Parkinson’s disease (PD), and Huntington’s disease (HD). Current preclinical data suggest stem cell transplantation is a potential treatment for these chronic conditions that lack effective long-term treatment options. Finding treatments with a wider therapeutic window and harnessing a disease-modifying approach will likely improve clinical outcomes. The overarching concept of stem cell therapy entails the use of immature cells, while key in recapitulating brain development and presents the challenge of young grafted cells forming neural circuitry with the mature host brain cells. To this end, exploring strategies designed to nurture graft-host integration will likely enhance the reconstruction of the elusive neural circuitry. Enriched environment (EE) and exercise facilitate stem cell graft-host reconstruction of neural circuitry. It may involve at least a two-pronged mechanism whereby EE and exercise create a conducive microenvironment in the host brain, allowing the newly transplanted cells to survive, proliferate, and differentiate into neural cells; vice versa, EE and exercise may also train the transplanted immature cells to learn the neurochemical, physiological, and anatomical signals in the brain towards better functional graft-host connectivity.
Collapse
Affiliation(s)
- Reed Berlet
- Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | | | | | - Cesar V. Borlongan
- Center of Excellence for Aging and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- *Correspondence: Cesar V. Borlongan,
| |
Collapse
|
5
|
Satani N, Parsha K, Savitz SI. Enhancing Stroke Recovery With Cellular Therapies. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00062-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
6
|
Liang CC, Shaw SW, Huang YH, Lee TH. Human amniotic fluid stem cells can improve cerebral vascular remodelling and neurological function after focal cerebral ischaemia in diabetic rats. J Cell Mol Med 2021; 25:10185-10196. [PMID: 34622573 PMCID: PMC8572791 DOI: 10.1111/jcmm.16956] [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: 04/15/2021] [Revised: 09/12/2021] [Accepted: 09/23/2021] [Indexed: 01/04/2023] Open
Abstract
Diabetes causes vascular injury and carries a high risk of ischaemic stroke. Human amniotic fluid stem cells (hAFSCs) can enhance cerebral vascular remodelling and have the potential to improve neurological function after stroke in diabetic rats. Five groups of female rats were examined: (1) normal control, (2) type 1 diabetic (T1DM) rats induced by streptozotocin injection (DM), (3) non‐DM rats receiving 60‐minute middle cerebral artery occlusion (MCAO), (4) T1DM rats receiving 60‐minute MCAO (DM + MCAO) and (5) T1DM rats receiving 60‐minute MCAO and injection with 5 × 106 hAFSCs at 3 h after MCAO (DM + MCAO + hAFSCs). Neurological function was examined before, and at 1, 7, 14, 21 and 28 days, and cerebral infarction volume and haemorrhage, cerebral vascular density, angiogenesis and inflammatory were examined at 7 and 28 days after MCAO. hAFSCs treatment caused a significant improvement of neurological dysfunction, infarction volume, blood‐brain barrier leakage, cerebral arterial density, vascular density and angiogenesis and a reduction of brain haemorrhage and inflammation compared with non‐treatment. Our results showed that the effect of hAFSCs treatment against focal cerebral ischaemia may act through the recovery of vascular remodelling and angiogenesis and the reduction of inflammation in ischaemic brain.
Collapse
Affiliation(s)
- Ching-Chung Liang
- Urogynecology Section, Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Steven W Shaw
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Obstetrics, Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei, Taiwan.,Prenatal Cell and Gene Therapy Group, Institute for Women's Health, University College London, London, UK
| | - Yung-Hsin Huang
- Urogynecology Section, Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Tsong-Hai Lee
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Stroke Center and Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| |
Collapse
|
7
|
Berlet R, Anthony S, Brooks B, Wang ZJ, Sadanandan N, Shear A, Cozene B, Gonzales-Portillo B, Parsons B, Salazar FE, Lezama Toledo AR, Monroy GR, Gonzales-Portillo JV, Borlongan CV. Combination of Stem Cells and Rehabilitation Therapies for Ischemic Stroke. Biomolecules 2021; 11:1316. [PMID: 34572529 PMCID: PMC8468342 DOI: 10.3390/biom11091316] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/14/2022] Open
Abstract
Stem cell transplantation with rehabilitation therapy presents an effective stroke treatment. Here, we discuss current breakthroughs in stem cell research along with rehabilitation strategies that may have a synergistic outcome when combined together after stroke. Indeed, stem cell transplantation offers a promising new approach and may add to current rehabilitation therapies. By reviewing the pathophysiology of stroke and the mechanisms by which stem cells and rehabilitation attenuate this inflammatory process, we hypothesize that a combined therapy will provide better functional outcomes for patients. Using current preclinical data, we explore the prominent types of stem cells, the existing theories for stem cell repair, rehabilitation treatments inside the brain, rehabilitation modalities outside the brain, and evidence pertaining to the benefits of combined therapy. In this review article, we assess the advantages and disadvantages of using stem cell transplantation with rehabilitation to mitigate the devastating effects of stroke.
Collapse
Affiliation(s)
- Reed Berlet
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd, North Chicago, IL 60064, USA;
| | - Stefan Anthony
- Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL 34211, USA;
| | - Beverly Brooks
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA; (B.B.); (Z.-J.W.)
| | - Zhen-Jie Wang
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA; (B.B.); (Z.-J.W.)
| | | | - Alex Shear
- University of Florida, 205 Fletcher Drive, Gainesville, FL 32611, USA;
| | - Blaise Cozene
- Tulane University, 6823 St. Charles Ave, New Orleans, LA 70118, USA;
| | | | - Blake Parsons
- Washington and Lee University, 204 W Washington St, Lexington, VA 24450, USA;
| | - Felipe Esparza Salazar
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan 52786, Mexico; (F.E.S.); (A.R.L.T.); (G.R.M.)
| | - Alma R. Lezama Toledo
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan 52786, Mexico; (F.E.S.); (A.R.L.T.); (G.R.M.)
| | - Germán Rivera Monroy
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan 52786, Mexico; (F.E.S.); (A.R.L.T.); (G.R.M.)
| | | | - Cesario V. Borlongan
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA; (B.B.); (Z.-J.W.)
- Center of Excellence for Aging and Brain Repair, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| |
Collapse
|
8
|
Alwjwaj M, Kadir RRA, Bayraktutan U. The secretome of endothelial progenitor cells: a potential therapeutic strategy for ischemic stroke. Neural Regen Res 2021; 16:1483-1489. [PMID: 33433461 PMCID: PMC8323700 DOI: 10.4103/1673-5374.303012] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Ischemic stroke continues to be a leading cause of mortality and morbidity in the world. Despite recent advances in the field of stroke medicine, thrombolysis with recombinant tissue plasminogen activator remains as the only pharmacological therapy for stroke patients. However, due to short therapeutic window (4.5 hours of stroke onset) and increased risk of hemorrhage beyond this point, each year globally less than 1% of stroke patients receive this therapy which necessitate the discovery of safe and efficacious therapeutics that can be used beyond the acute phase of stroke. Accumulating evidence indicates that endothelial progenitor cells (EPCs), equipped with an inherent capacity to migrate, proliferate and differentiate, may be one such therapeutics. However, the limited availability of EPCs in peripheral blood and early senescence of few isolated cells in culture conditions adversely affect their application as effective therapeutics. Given that much of the EPC-mediated reparative effects on neurovasculature is realized by a wide range of biologically active substances released by these cells, it is possible that EPC-secretome may serve as an important therapeutic after an ischemic stroke. In light of this assumption, this review paper firstly discusses the main constituents of EPC-secretome that may exert the beneficial effects of EPCs on neurovasculature, and then reviews the currently scant literature that focuses on its therapeutic capacity.
Collapse
Affiliation(s)
- Mansour Alwjwaj
- Stroke, Division of Clinical Neuroscience, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Rais Reskiawan A Kadir
- Stroke, Division of Clinical Neuroscience, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Ulvi Bayraktutan
- Stroke, Division of Clinical Neuroscience, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK
| |
Collapse
|
9
|
Sitruk-Ware R, Bonsack B, Brinton R, Schumacher M, Kumar N, Lee JY, Castelli V, Corey S, Coats A, Sadanandan N, Gonzales-Portillo B, Heyck M, Shear A, Blaise C, Zhang H, Sheyner M, García-Sánchez J, Navarro L, El-Etr M, De Nicola AF, Borlongan CV. Progress in progestin-based therapies for neurological disorders. Neurosci Biobehav Rev 2020; 122:38-65. [PMID: 33359391 DOI: 10.1016/j.neubiorev.2020.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/26/2020] [Accepted: 12/12/2020] [Indexed: 12/16/2022]
Abstract
Hormone therapy, primarily progesterone and progestins, for central nervous system (CNS) disorders represents an emerging field of regenerative medicine. Following a failed clinical trial of progesterone for traumatic brain injury treatment, attention has shifted to the progestin Nestorone for its ability to potently and selectively transactivate progesterone receptors at relatively low doses, resulting in robust neurogenetic, remyelinating, and anti-inflammatory effects. That CNS disorders, including multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), spinal cord injury (SCI), and stroke, develop via demyelinating, cell death, and/or inflammatory pathological pathways advances Nestorone as an auspicious candidate for these disorders. Here, we assess the scientific and clinical progress over decades of research into progesterone, progestins, and Nestorone as neuroprotective agents in MS, ALS, SCI, and stroke. We also offer recommendations for optimizing timing, dosage, and route of the drug regimen, and identifying candidate patient populations, in advancing Nestorone to the clinic.
Collapse
Affiliation(s)
| | - Brooke Bonsack
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | | | | | | | - Jea-Young Lee
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Vanessa Castelli
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Sydney Corey
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Alexandreya Coats
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Nadia Sadanandan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Bella Gonzales-Portillo
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Matt Heyck
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Alex Shear
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Cozene Blaise
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Henry Zhang
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Michael Sheyner
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Julián García-Sánchez
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Lisset Navarro
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | | | | | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| |
Collapse
|
10
|
Bagheri-Mohammadi S. Protective effects of mesenchymal stem cells on ischemic brain injury: therapeutic perspectives of regenerative medicine. Cell Tissue Bank 2020; 22:249-262. [PMID: 33231840 DOI: 10.1007/s10561-020-09885-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 11/12/2020] [Indexed: 12/19/2022]
Abstract
Cerebral ischemic injury as the main manifestation of stroke can occur in stroke patients (70-80%). Nowadays, the main therapeutic strategy used for ischemic brain injury treatment aims to achieve reperfusion, neuroprotection, and neurorecovery. Also, angiogenesis as a therapeutic approach maybe represents a promising tool to enhance the prognosis of cerebral ischemic stroke. Unfortunately, although many therapeutic approaches as a life-saving gateway for cerebral ischemic injuries like pharmacotherapy and surgical treatments are widely used, they all fail to restore or regenerate damaged neurons in the brain. So, the suitable therapeutic approach would focus on regenerating the lost cells and restore the normal function of the brain. Currently, stem cell-based regenerative medicine introduced a new paradigm approach in cerebral ischemic injuries treatment. Today, in experimental researches, different types of stem cells such as mesenchymal stem cells have been applied. Therefore, stem cell-based regenerative medicine provides the opportunity to inquire and develop a more effective and safer therapeutic approach with the capability to produce and regenerate new neurons in damaged tissues.
Collapse
Affiliation(s)
- Saeid Bagheri-Mohammadi
- Department of Physiology and Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Physiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
- Departments of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
11
|
Abstract
Stroke remains a major unmet clinical need that warrants novel therapies. Following an ischemic insult, the cerebral vasculature secretes inflammatory molecules, creating the stroke vasculome profile. The present study evaluated the therapeutic effects of endothelial cells on the inflammation-associated stroke vasculome. qRT-PCR analysis revealed that specific inflammation-related vasculome genes BRM, IκB, Foxf1, and ITIH-5 significantly upregulated by oxygen glucose deprivation (OGD. Interestingly, co-culture of human endothelial cells (HEN6) with human endothelial cells (EPCs) during OGD significantly blocked the elevations of BRM, IκB, and Foxf1, but not ITIH-5. Next, employing the knockdown/antisense technology, silencing the inflammation-associated stroke vasculome gene, IκB, as opposed to scrambled knockdown, blocked the EPC-mediated protection of HEN6 against OGD. In vivo, stroke animals transplanted with intracerebral human EPCs (300,000 cells) into the striatum and cortex 4 h post ischemic stroke displayed significant behavioral recovery up to 30 days post-transplantation compared to vehicle-treated stroke animals. At 7 days post-transplantation, quantification of the fluorescent staining intensity in the cortex and striatum revealed significant upregulation of the endothelial marker RECA1 and a downregulation of the stroke-associated vasculome BRM, IKB, Foxf1, ITIH-5 and PMCA2 in the ipsilateral side of cortex and striatum of EPC-transplanted stroke animals relative to vehicle-treated stroke animals. Altogether, these results demonstrate that EPCs exert therapeutic effects in experimental stroke possibly by modulating the inflammation-plagued vasculome.
Collapse
|
12
|
Hu H, Zhao Q, Liu X, Yan T. Human umbilical cord blood cells rescued traumatic brain injury-induced cardiac and neurological deficits. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:278. [PMID: 32355722 PMCID: PMC7186665 DOI: 10.21037/atm.2020.03.52] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background Traumatic brain injury (TBI) evokes neurological deficits and induces cardiac dysfunction. Treatment with human umbilical cord blood cells (HUCBCs) represents a potential therapeutic strategy for TBI-induced neurological deficits. The present study aimed to determine whether HUCBCs could ameliorate the cardiac dysfunction and neurological deficits induced by TBI. Methods Adult male C57BL/6J mice were subjected to controlled cortical impact (CCI)-induced TBI and were treated with either HUCBCs (1×106) or phosphate-buffered saline (PBS), via tail vein injections, 3 days after TBI. Neurological and cognitive functions were subsequently evaluated at multiple time points after TBI and cardiac function was assessed by echocardiography 3 and 30 days after TBI. Brain and heart tissues were paraffin-embedded 30 days after TBI. Hematoxylin and eosin (H&E) staining was performed on brain tissue sections to calculate the brain damage volume, and Picro Sirius Red (PSR) staining was performed on heart tissue sections to evaluate myocardial fibrosis. Terminal deoxynucleotide transferase dUTP nick end labeling (TUNEL) staining was employed to assess cell apoptosis 30 days after TBI. Transforming growth factor-beta (TGF-β) and NADPH oxidase-2 (NOX2) levels were assessed to evaluate inflammation and oxidative stress levels 30 days after TBI. Results TBI elicited acute and chronic cardiac deficits, identified by decreased left ventricular ejection fraction (LVEF) and fractional shortening (LVFS) values 3 and 30 days after TBI, in addition to neurological and cognitive deficits. TBI mice treated with HUCBCs exhibited enhanced LVEF and FS values 30 days after TBI compared with untreated TBI controls. HUCBC treatment significantly improved neurological and cognitive functions and reduced cardiomyocyte apoptosis, inflammatory response, oxidative stress, and cardiac fibrosis in heart tissues 30 days after TBI. Conclusions TBI induced both neurological deficits and cardiac dysfunction in mice, which were ameliorated by HUCBC treatment. The anti-inflammatory activities of HUCBCs may contribute to these observed therapeutic effects.
Collapse
Affiliation(s)
- Haotian Hu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma, Neurorepair, and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Qiang Zhao
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma, Neurorepair, and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Xiaoxuan Liu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma, Neurorepair, and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Tao Yan
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma, Neurorepair, and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| |
Collapse
|
13
|
Corey S, Bonsack B, Heyck M, Shear A, Sadanandan N, Zhang H, Borlongan CV. Harnessing the anti-inflammatory properties of stem cells for transplant therapy in hemorrhagic stroke. BRAIN HEMORRHAGES 2020; 1:24-33. [PMID: 34056567 PMCID: PMC8158660 DOI: 10.1016/j.hest.2019.12.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hemorrhagic stroke is a global health crisis plagued by neuroinflammation in the acute and chronic phases. Neuroinflammation approximates secondary cell death, which in turn robustly contributes to stroke pathology. Both the physiological and behavioral symptoms of stroke correlate with various inflammatory responses in animal and human studies. That slowing the secondary cell death mediated by this inflammation may attenuate stroke pathology presents a novel treatment strategy. To this end, experimental therapies employing stem cell transplants support their potential for neuroprotection and neuroregeneration after hemorrhagic stroke. In this review, we evaluate experiments using different types of stem cell transplants as treatments for stroke-induced neuroinflammation. We also update this emerging area by examining recent preclinical and clinical trials that have deployed these therapies. While further investigations are warranted to solidify their therapeutic profile, the reviewed studies largely posit stem cells as safe and potent biologics for stroke, specifically owing to their mode of action for sequestering neuroinflammation and promoting neuroregenerative processes.
Collapse
Affiliation(s)
- Sydney Corey
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Brooke Bonsack
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Matt Heyck
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Alex Shear
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Nadia Sadanandan
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Henry Zhang
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| |
Collapse
|
14
|
Tuazon JP, Sitruk-Ware R, Borlongan CV. Reprint of: Beyond contraception and hormone replacement therapy: Advancing Nestorone to a neuroprotective drug in the clinic. Brain Res 2019; 1719:285-287. [PMID: 31213288 DOI: 10.1016/j.brainres.2019.05.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Neurological diseases such as ischemic stroke can be debilitating and have limited treatments available. The progestin Nestorone® (segesterone acetate) has been evaluated for use in birth control and hormone replacement therapy due to its potency and high affinity for the progesterone receptor. Interestingly, Nestorone also exerts neuroprotection in animals afflicted with various central nervous system diseases, including stroke, which implicates its potential for treating these maladies in clinical settings. In fact, a recent Brain Research paper by Tanaka and colleagues demonstrates Nestorone's ability to reduce infarct sizes and preclude functional impairments in rats subjected to ischemic stroke. This commentary highlights Nestorone's properties as a progestin, its neuroprotective capabilities in animal studies, and how the Tanaka team's findings and previous clinical trials contribute to Nestorone's translation into a therapeutic agent for stroke and other neurological diseases.
Collapse
Affiliation(s)
- Julian P Tuazon
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL 33612, USA
| | - Regine Sitruk-Ware
- Population Council, Center for Biomedical Research, 1230 York Avenue, New York, NY 10065, USA
| | - Cesario V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL 33612, USA.
| |
Collapse
|
15
|
Neal EG, Acosta SA, Kaneko Y, Ji X, Borlongan CV. Regulatory T-cells within bone marrow-derived stem cells actively confer immunomodulatory and neuroprotective effects against stroke. J Cereb Blood Flow Metab 2019; 39:1750-1758. [PMID: 29569981 PMCID: PMC6727132 DOI: 10.1177/0271678x18766172] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/15/2018] [Accepted: 02/20/2018] [Indexed: 12/15/2022]
Abstract
Regulatory T-cells (Tregs) may exert a neuroprotective effect on ischemic stroke by inhibiting both inflammation and effector T-cell activation. Transplantation of human bone marrow-derived stem cells (BMSCs) in ischemic stroke affords neuroprotection that results in part from the cells' anti-inflammatory property. However, the relationship between Tregs and BMSCs in treatment of ischemic stroke has not been fully elucidated. Here, we tested the hypothesis that Tregs within the BMSCs represent active mediators of immunomodulation and neuroprotection in experimental stroke. Primary rat neuronal cells were subjected to an oxygen-glucose deprivation and reperfusion (OGD/R) condition. The cells were re-perfused and co-cultured with Tregs and/or BMSCs. We detected a minority population of Tregs within BMSCs with both immunocytochemistry (ICC) and flow cytometry identifying cells expressing phenotypic markers of CD4, CD25, and FoxP3 protein. BMSCs with the native population of Tregs conferred maximal neuroprotection compared to the treatment conditions containing 0%, 10%, and 100% relative ratio Tregs. Increasing the Treg population resulted in increased IL6 secretion and decreased FGF-β secretion by BMSCs. This study shows that a minority population of Tregs exists within the therapeutic BMSC population, which serves as robust mediators of the immunomodulatory and neuroprotective effect provided by BMSC transplantation.
Collapse
Affiliation(s)
- Elliot G Neal
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, USF Morsani College of Medicine, Tampa, FL, USA
| | - Sandra A Acosta
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, USF Morsani College of Medicine, Tampa, FL, USA
| | - Yuji Kaneko
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, USF Morsani College of Medicine, Tampa, FL, USA
| | - Xunming Ji
- Cerebrovascular Research Center, XuanWu Hospital, Capital Medical University, Beijing, China
| | - Cesario V Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, USF Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
16
|
Abe Y, Ochiai D, Masuda H, Sato Y, Otani T, Fukutake M, Ikenoue S, Miyakoshi K, Okano H, Tanaka M. In Utero Amniotic Fluid Stem Cell Therapy Protects Against Myelomeningocele via Spinal Cord Coverage and Hepatocyte Growth Factor Secretion. Stem Cells Transl Med 2019; 8:1170-1179. [PMID: 31407874 PMCID: PMC6811697 DOI: 10.1002/sctm.19-0002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 07/02/2019] [Indexed: 12/13/2022] Open
Abstract
Despite the poor prognosis associated with myelomeningocele (MMC), the options for prenatal treatments are still limited. Recently, fetal cellular therapy has become a new option for treating birth defects, although the therapeutic effects and mechanisms associated with such treatments remain unclear. The use of human amniotic fluid stem cells (hAFSCs) is ideal with respect to immunoreactivity and cell propagation. The prenatal diagnosis of MMC during early stages of pregnancy could allow for the ex vivo proliferation and modulation of autologous hAFSCs for use in utero stem cell therapy. Therefore, we investigated the therapeutic effects and mechanisms of hAFSCs‐based treatment for fetal MMC. hAFSCs were isolated as CD117‐positive cells from the amniotic fluid of 15‐ to 17‐week pregnant women who underwent amniocentesis for prenatal diagnosis and consented to this study. Rat dams were exposed to retinoic acid to induce fetal MMC and were subsequently injected with hAFSCs in each amniotic cavity. We measured the exposed area of the spinal cord and hepatocyte growth factor (HGF) levels at the lesion. The exposed spinal area of the hAFSC‐treated group was significantly smaller than that of the control group. Immunohistochemical analysis demonstrated a reduction in neuronal damage such as neurodegeneration and astrogliosis in the hAFSC‐treated group. Additionally, in lesions of the hAFSC‐treated group, HGF expression was upregulated and HGF‐positive hAFSCs were identified, suggesting that these cells migrated to the lesion and secreted HGF to suppress neuronal damage and induce neurogenesis. Therefore, in utero hAFSC therapy could become a novel strategy for fetal MMC. stem cells translational medicine2019;8:1170–1179
Collapse
Affiliation(s)
- Yushi Abe
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Daigo Ochiai
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Hirotaka Masuda
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Yu Sato
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Toshimitsu Otani
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Marie Fukutake
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Satoru Ikenoue
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kei Miyakoshi
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Mamoru Tanaka
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
17
|
Tuazon JP, Castelli V, Borlongan CV. Drug-like delivery methods of stem cells as biologics for stroke. Expert Opin Drug Deliv 2019; 16:823-833. [PMID: 31311344 DOI: 10.1080/17425247.2019.1645116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Stem cell therapy is an experimental treatment for brain disorders. Although a cellular product, stem cells can be classified as biologics based on the cells' secretion of therapeutic substances. Treatment with stem cell biologics may appeal to stroke because of the secondary cell death mechanisms, especially neuroinflammation, that are rampant from the onset and remain elevated during the progressive phase of the disease requiring multi-pronged biological targets to effectively abrogate the neurodegenerative pathology. However, the optimal delivery methods, among other logistical approaches (i.e. cell doses and timing of intervention), for stem cell therapy will need to be refined before stem cell biologics can be successfully utilized for stroke in large scale clinical trials. Areas covered: In this review, we discuss how the innate qualities of stem cells characterize them as biologics, how stem cell transplantation may be an ideal treatment for stroke, and the various routes of stem cell administration that have been employed in various preclinical and clinical investigations. Expert opinion: There is a need to optimize the delivery of stem cell biologics for stroke in order to guide the safe and effective translation of this therapy from the laboratory to the clinic.
Collapse
Affiliation(s)
- Julian P Tuazon
- a Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine , Tampa , FL , USA
| | - Vanessa Castelli
- a Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine , Tampa , FL , USA
| | - Cesar V Borlongan
- a Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine , Tampa , FL , USA
| |
Collapse
|
18
|
Sibov TT, Pavon LF, Cabral FR, Cunha IF, de Oliveira DM, de Souza JG, Marti LC, da Cruz EF, Malheiros JM, Paiva FF, Tannús A, de Oliveira SM, da Costa MDS, Dastoli PA, Mendonça JN, de Toledo SRC, Malheiros SMF, de Paiva Neto MA, Rego NBB, Moron AF, Cavalheiro S. Intravenous Grafts of Human Amniotic Fluid-Derived Stem Cells Reduce Behavioral Deficits in Experimental Ischemic Stroke. Cell Transplant 2019; 28:1306-1320. [PMID: 31161782 PMCID: PMC6767884 DOI: 10.1177/0963689719854342] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Amniotic fluid has been investigated as new cell source for stem cells in the development
of future cell-based transplantation. This study reports isolation of viable human
amniotic fluid-derived stem cells, labeled with multimodal iron oxide nanoparticles, and
its effect on focal cerebral ischemia–reperfusion injury in Wistar rats. Middle cerebral
artery occlusion of 60 min followed by reperfusion for 1 h, 6 h, and 24 h was employed in
the present study to produce ischemia and reperfusion-induced cerebral injury in rats.
Tests were employed to assess the functional outcome of the sensorimotor center activity
in the brain, through a set of modified neurological severity scores used to assess motor
and exploratory capacity 24 h, 14, and 28 days after receiving cellular therapy via tail
vein. In our animal model of stroke, transplanted cells migrated to the ischemic focus,
infarct volume decreased, and motor deficits improved. Therefore, we concluded that these
cells appear to have beneficial effects on the ischemic brain, possibly based on their
ability to enhance endogenous repair mechanisms.
Collapse
Affiliation(s)
- Tatiana Taís Sibov
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Lorena Favaro Pavon
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Francisco Romero Cabral
- Hospital Israelita Albert Einstein (HIAE), Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| | - Ivone Farias Cunha
- Hospital Israelita Albert Einstein (HIAE), Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| | | | | | - Luciana Cavalheiro Marti
- Hospital Israelita Albert Einstein (HIAE), Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| | - Edgar Ferreira da Cruz
- Department of Medicine, Discipline of Nephrology, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | | | - Fernando F Paiva
- São Carlos Institute of Physics, São Paulo University, São Paulo, Brazil
| | - Alberto Tannús
- São Carlos Institute of Physics, São Paulo University, São Paulo, Brazil
| | | | - Marcos Devanir Silva da Costa
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Patrícia A Dastoli
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Jardel N Mendonça
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Silvia Regina Caminada de Toledo
- Pediatrics Oncology Institute, GRAACC (Grupo de Apoio ao Adolescente e a Criança com Câncer), Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Suzana M Fleury Malheiros
- Hospital Israelita Albert Einstein (HIAE), Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil.,Department of Neuro-Oncology, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Manoel Antonio de Paiva Neto
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Nelma Bastos Bezerra Rego
- Department of Obstetrics, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Antônio Fernandes Moron
- Department of Obstetrics, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Sérgio Cavalheiro
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina-Universidade Federal de São Paulo (EPM-UNIFESP), São Paulo, Brazil
| |
Collapse
|
19
|
Nguyen H, Zarriello S, Coats A, Nelson C, Kingsbury C, Gorsky A, Rajani M, Neal EG, Borlongan CV. Stem cell therapy for neurological disorders: A focus on aging. Neurobiol Dis 2019; 126:85-104. [PMID: 30219376 PMCID: PMC6650276 DOI: 10.1016/j.nbd.2018.09.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/04/2018] [Accepted: 09/11/2018] [Indexed: 02/07/2023] Open
Abstract
Age-related neurological disorders continue to pose a significant societal and economic burden. Aging is a complex phenomenon that affects many aspects of the human body. Specifically, aging can have detrimental effects on the progression of brain diseases and endogenous stem cells. Stem cell therapies possess promising potential to mitigate the neurological symptoms of such diseases. However, aging presents a major obstacle for maximum efficacy of these treatments. In this review, we discuss current preclinical and clinical literature to highlight the interactions between aging, stem cell therapy, and the progression of major neurological disease states such as Parkinson's disease, Huntington's disease, stroke, traumatic brain injury, amyotrophic lateral sclerosis, multiple sclerosis, and multiple system atrophy. We raise important questions to guide future research and advance novel treatment options.
Collapse
Affiliation(s)
- Hung Nguyen
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Sydney Zarriello
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Alexandreya Coats
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Cannon Nelson
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Chase Kingsbury
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Anna Gorsky
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Mira Rajani
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Elliot G Neal
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA.
| |
Collapse
|
20
|
Pischiutta F, Sammali E, Parolini O, Carswell HVO, Zanier ER. Placenta-Derived Cells for Acute Brain Injury. Cell Transplant 2019; 27:151-167. [PMID: 29562781 PMCID: PMC6434489 DOI: 10.1177/0963689717732992] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Acute brain injury resulting from ischemic/hemorrhagic or traumatic damage is one of the leading causes of mortality and disability worldwide and is a significant burden to society. Neuroprotective options to counteract brain damage are very limited in stroke and traumatic brain injury (TBI). Given the multifaceted nature of acute brain injury and damage progression, several therapeutic targets may need to be addressed simultaneously to interfere with the evolution of the injury and improve the patient’s outcome. Stem cells are ideal candidates since they act on various mechanisms of protection and repair, improving structural and functional outcomes after experimental stroke or TBI. Stem cells isolated from placenta offer advantages due to their early embryonic origin, ease of procurement, and ethical acceptance. We analyzed the evidence for the beneficial effects of placenta-derived stem cells in acute brain injury, with the focus on experimental studies of TBI and stroke, the engineering strategies pursued to foster cell potential, and characterization of the bioactive molecules secreted by placental cells, known as their secretome, as an alternative cell-free strategy. Results from the clinical application of placenta-derived stem cells for acute brain injury and ongoing clinical trials are summarily discussed.
Collapse
Affiliation(s)
- Francesca Pischiutta
- 1 Department of Neuroscience, Laboratory of Acute Brain Injury and Therapeutic Strategies, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Eliana Sammali
- 1 Department of Neuroscience, Laboratory of Acute Brain Injury and Therapeutic Strategies, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy.,2 Department of Cerebrovascular Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Ornella Parolini
- 3 Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy.,4 Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Hilary V O Carswell
- 5 Strathclyde Institute of Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow, United Kingdom
| | - Elisa R Zanier
- 1 Department of Neuroscience, Laboratory of Acute Brain Injury and Therapeutic Strategies, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| |
Collapse
|
21
|
Tuazon JP, Sitruk-Ware R, Borlongan CV. Beyond contraception and hormone replacement therapy: Advancing Nestorone to a neuroprotective drug in the clinic. Brain Res 2019; 1704:161-163. [DOI: 10.1016/j.brainres.2018.10.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 10/11/2018] [Indexed: 01/17/2023]
|
22
|
Loukogeorgakis SP, De Coppi P. Concise Review: Amniotic Fluid Stem Cells: The Known, the Unknown, and Potential Regenerative Medicine Applications. Stem Cells 2018; 35:1663-1673. [PMID: 28009066 DOI: 10.1002/stem.2553] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 09/07/2016] [Accepted: 10/01/2016] [Indexed: 12/19/2022]
Abstract
The amniotic fluid has been identified as an untapped source of cells with broad potential, which possess immunomodulatory properties and do not have the ethical and legal limitations of embryonic stem cells. CD117(c-Kit)+ cells selected from amniotic fluid have been shown to differentiate into cell lineages representing all three embryonic germ layers without generating tumors, making them ideal candidates for regenerative medicine applications. Moreover, their ability to engraft in injured organs and modulate immune and repair responses of host tissues, suggest that transplantation of such cells may be useful for the treatment of various degenerative and inflammatory diseases. Although significant questions remain regarding the origin, heterogeneous phenotype, and expansion potential of amniotic fluid stem cells, evidence to date supports their potential role as a valuable stem cell source for the field of regenerative medicine. Stem Cells 2017;35:1663-1673.
Collapse
Affiliation(s)
- Stavros P Loukogeorgakis
- Stem Cells and Regenerative Medicine Section, Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom
| | - Paolo De Coppi
- Stem Cells and Regenerative Medicine Section, Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
23
|
Human Amniotic Fluid Stem Cells: Therapeutic Potential for Perinatal Patients with Intractable Neurological Disease. Keio J Med 2018. [PMID: 29515049 DOI: 10.2302/kjm.2017-0019-ir] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mesenchymal stem cells (MSCs) have generated great interest in the fields of regenerative medicine and immunotherapy because of their unique biological properties. Among MSCs, amniotic fluid stem cells (AFS) have a number of characteristics that make them attractive candidates for tissue engineering and cell replacement strategies, particularly for perinatal medicine. If various neonatal conditions, including birth asphyxia, preterm birth, and congenital abnormalities, which result in long-lasting severe impairments, could be predicted during pregnancy, it would allow collection of small samples of amniotic fluid cells by amniocentesis. In vitro culture of these autologous AFS during pregnancy would make them available for use soon after birth. Hypoxic-ischemic encephalopathy (HIE) and myelomeningocele (MMC) are neonatal conditions that cause permanent neurological disability, for which the treatment options are extremely limited. Experiments using animal models of HIE and MMC and human clinical trials have demonstrated that MSCs, including AFS, have beneficial effects on the central nervous system through paracrine influences, indicating that autologous AFS treatment may be applicable for intractable neurological diseases, including HIE and MMC, during the perinatal period. In this review, we focus on recent research related to the therapeutic potential of AFS for perinatal neurological diseases such as HIE and MMC.
Collapse
|
24
|
Napoli E, Borlongan CV. Stem Cell Recipes of Bone Marrow and Fish: Just What the Stroke Doctors Ordered. Stem Cell Rev Rep 2017; 13:192-197. [PMID: 28064388 DOI: 10.1007/s12015-016-9716-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Stem cell therapy for stroke has advanced from the laboratory to the clinic, but remains as an experimental treatment. Two lines of transplant regimens have emerged, namely the "early bird" peripheral injections in subacute stroke patients and the "late night" direct intracerebral treatments in chronic stroke patients. Autologous bone marrow-derived stem cells, which only required minimal manipulations during graft cell preparation, gained fast-track entry into the clinic, while gene modified stem cells necessitated overcoming more stringent regulatory criteria before they were approved for clinical use. Safety of the stem cell therapy can be declared from these clinical trials, but efficacy warrants further investigations. Here, we offer insights into the translation of cell therapy from the laboratory to the clinic, in the hopes that highlighting the lessons we learned from this experience will guide the optimization of functional outcomes of future clinical trials of stem cell therapy for stroke.
Collapse
Affiliation(s)
- Eleonora Napoli
- Department of Molecular Biosciences, University of California Davis, Davis, CA, 95616, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, 33612, USA.
| |
Collapse
|
25
|
Stem cell therapy for abrogating stroke-induced neuroinflammation and relevant secondary cell death mechanisms. Prog Neurobiol 2017; 158:94-131. [PMID: 28743464 DOI: 10.1016/j.pneurobio.2017.07.004] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/13/2022]
Abstract
Ischemic stroke is a leading cause of death worldwide. A key secondary cell death mechanism mediating neurological damage following the initial episode of ischemic stroke is the upregulation of endogenous neuroinflammatory processes to levels that destroy hypoxic tissue local to the area of insult, induce apoptosis, and initiate a feedback loop of inflammatory cascades that can expand the region of damage. Stem cell therapy has emerged as an experimental treatment for stroke, and accumulating evidence supports the therapeutic efficacy of stem cells to abrogate stroke-induced inflammation. In this review, we investigate clinically relevant stem cell types, such as hematopoietic stem cells (HSCs), mesenchymal stem cells (MSCs), endothelial progenitor cells (EPCs), very small embryonic-like stem cells (VSELs), neural stem cells (NSCs), extraembryonic stem cells, adipose tissue-derived stem cells, breast milk-derived stem cells, menstrual blood-derived stem cells, dental tissue-derived stem cells, induced pluripotent stem cells (iPSCs), teratocarcinoma-derived Ntera2/D1 neuron-like cells (NT2N), c-mycER(TAM) modified NSCs (CTX0E03), and notch-transfected mesenchymal stromal cells (SB623), comparing their potential efficacy to sequester stroke-induced neuroinflammation and their feasibility as translational clinical cell sources. To this end, we highlight that MSCs, with a proven track record of safety and efficacy as a transplantable cell for hematologic diseases, stand as an attractive cell type that confers superior anti-inflammatory effects in stroke both in vitro and in vivo. That stem cells can mount a robust anti-inflammatory action against stroke complements the regenerative processes of cell replacement and neurotrophic factor secretion conventionally ascribed to cell-based therapy in neurological disorders.
Collapse
|
26
|
Antonucci I, Crowley MG, Stuppia L. Amniotic fluid stem cell models: A tool for filling the gaps in knowledge for human genetic diseases. Brain Circ 2017; 3:167-174. [PMID: 30276320 PMCID: PMC6057697 DOI: 10.4103/bc.bc_23_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 09/06/2017] [Accepted: 09/12/2017] [Indexed: 12/21/2022] Open
Abstract
Induced pluripotent stem (iPS) cells have attracted attention in recent years as a model of human genetic diseases. Starting from the diseased somatic cells isolated from an affected patient, iPS cells can be created and subsequently differentiated into various cell types that can be used to gain a better understanding of the disease at a cellular and molecular level. There are limitations of iPS cell generation, however, due to low efficiency, high costs, and lengthy protocols. The use of amniotic fluid stem cells (AFS) presents a worthy alternative as a stem cell source for modeling of human genetic diseases. Prenatal identification of chromosomal or Mendelian diseases may require the collection of amniotic fluid which is not only useful for the sake of diagnosis but also from this, AFS cells can be isolated and cultured. Since AFS cells show some characteristics of pluripotency, having the capacity to differentiate into various cell types derived from all three germ layers in vitro, they are a well-suited model for investigations regarding alterations in the molecular biology of a cell due to a specific genetic disease. This readily accessible source of stem cells can replace the necessity for generating iPS cells. Here, we expand on the applicability and importance of AFS cells as a model for discovery in the field of human genetic disease research. This paper is a review article. Referred literature in this paper has been listed in the references section. The data sets supporting the conclusions of this article are available online by searching various databases, including PubMed. Some original points in this article come from the laboratory practice in our research center and the authors’ experiences.
Collapse
Affiliation(s)
- Ivana Antonucci
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, Annunzio University, Chieti-Pescara, Italy
| | - Marci G Crowley
- Center of Excellence for Aging and Brain Repair, University of South Florida, 12901, USA
| | - Liborio Stuppia
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, Annunzio University, Chieti-Pescara, Italy
| |
Collapse
|
27
|
Lee JY, Xu K, Nguyen H, Guedes VA, Borlongan CV, Acosta SA. Stem Cell-Induced Biobridges as Possible Tools to Aid Neuroreconstruction after CNS Injury. Front Cell Dev Biol 2017; 5:51. [PMID: 28540289 PMCID: PMC5424542 DOI: 10.3389/fcell.2017.00051] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 04/21/2017] [Indexed: 12/12/2022] Open
Abstract
Notch-induced mesenchymal stromal cells (MSCs) mediate a distinct mechanism of repair after brain injury by forming a biobridge that facilitates biodistribution of host cells from a neurogenic niche to the area of injury. We have observed the biobridge in an area between the subventricular zone and the injured cortex using immunohistochemistry and laser capture. Cells in the biobridge express high levels of extracellular matrix metalloproteinases (MMPs), specifically MMP-9, which co-localized with a trail of MSCs graft. The transplanted stem cells then become almost undetectable, being replaced by newly recruited host cells. This stem cell-paved biobridge provides support for distal migration of host cells from the subventricular zone to the site of injury. Biobridge formation by transplanted stem cells seems to have a fundamental role in initiating endogenous repair processes. Two major stem cell-mediated repair mechanisms have been proposed thus far: direct cell replacement by transplanted grafts and bystander effects through the secretion of trophic factors including fibroblast growth factor 2 (FGF-2), epidermal growth factor (EGF), stem cell factor (SCF), erythropoietin, and brain-derived neurotrophic factor (BDNF) among others. This groundbreaking observation of biobridge formation by transplanted stem cells represents a novel mechanism for stem cell mediated brain repair. Future studies on graft-host interaction will likely establish biobridge formation as a fundamental mechanism underlying therapeutic effects of stem cells and contribute to the scientific pursuit of developing safe and efficient therapies not only for traumatic brain injury but also for other neurological disorders. The aim of this review is to hypothetically extend concepts related to the formation of biobridges in other central nervous system disorders.
Collapse
Affiliation(s)
- Jea Y Lee
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Kaya Xu
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Hung Nguyen
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Vivian A Guedes
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Sandra A Acosta
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| |
Collapse
|
28
|
Tajiri N, Quach DM, Kaneko Y, Wu S, Lee D, Lam T, Hayama KL, Hazel TG, Johe K, Wu MC, Borlongan CV. NSI-189, a small molecule with neurogenic properties, exerts behavioral, and neurostructural benefits in stroke rats. J Cell Physiol 2017; 232:2731-2740. [PMID: 28181668 PMCID: PMC5518191 DOI: 10.1002/jcp.25847] [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: 01/19/2017] [Accepted: 02/07/2017] [Indexed: 12/26/2022]
Abstract
Enhancing neurogenesis may be a powerful stroke therapy. Here, we tested in a rat model of ischemic stroke the beneficial effects of NSI-189, an orally active, new molecular entity (mol. wt. 366) with enhanced neurogenic activity, and indicated as an anti-depressant drug in a clinical trial (Fava et al., , Molecular Psychiatry, DOI: 10.1038/mp.2015.178) and being tested in a Phase 2 efficacy trial (ClinicalTrials.gov, , ClinicalTrials.gov Identifier: NCT02695472) for treatment of major depression. Oral administration of NSI-189 in adult Sprague-Dawley rats starting at 6 hr after middle cerebral artery occlusion, and daily thereafter over the next 12 weeks resulted in significant amelioration of stroke-induced motor and neurological deficits, which was maintained up to 24 weeks post-stroke. Histopathological assessment of stroke brains from NSI-189-treated animals revealed significant increments in neurite outgrowth as evidenced by MAP2 immunoreactivity that was prominently detected in the hippocampus and partially in the cortex. These results suggest NSI-189 actively stimulated remodeling of the stroke brain. Parallel in vitro studies further probed this remodeling process and demonstrated that oxygen glucose deprivation and reperfusion (OGD/R) initiated typical cell death processes, which were reversed by NSI-189 treatment characterized by significant attenuation of OGD/R-mediated hippocampal cell death and increased Ki67 and MAP2 expression, coupled with upregulation of neurogenic factors such as BDNF and SCF. These findings support the use of oral NSI-189 as a therapeutic agent well beyond the initial 6-hr time window to accelerate and enhance the overall functional improvement in the initial 6 months post stroke.
Collapse
Affiliation(s)
- Naoki Tajiri
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University South Florida College of Medicine, Tampa, Florida
| | | | - Yuji Kaneko
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University South Florida College of Medicine, Tampa, Florida
| | | | - David Lee
- Neuralstem, Inc., Rockville, Maryland
| | - Tina Lam
- Neuralstem, Inc., Rockville, Maryland
| | | | | | - Karl Johe
- Neuralstem, Inc., Rockville, Maryland
| | | | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University South Florida College of Medicine, Tampa, Florida
| |
Collapse
|
29
|
Garbuzova-Davis S, Haller E, Lin R, Borlongan CV. Intravenously Transplanted Human Bone Marrow Endothelial Progenitor Cells Engraft Within Brain Capillaries, Preserve Mitochondrial Morphology, and Display Pinocytotic Activity Toward Blood-Brain Barrier Repair in Ischemic Stroke Rats. Stem Cells 2017; 35:1246-1258. [PMID: 28142208 DOI: 10.1002/stem.2578] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 01/05/2017] [Accepted: 01/12/2017] [Indexed: 01/01/2023]
Abstract
Stroke is a life-threatening disease with limited therapeutic options. Cell therapy has emerged as an experimental stroke treatment. Blood-brain barrier (BBB) impairment is a key pathological manifestation of ischemic stroke, and barrier repair is an innovative target for neurorestoration in stroke. Here, we evaluated via electron microscopy the ability of transplanted human bone marrow endothelial progenitor cells (hBMEPCs) to repair the BBB in adult Sprague-Dawley rats subjected to transient middle cerebral artery occlusion (tMCAO). β-galactosidase prelabeled hBMEPCs were intravenously transplanted 48 hours post-tMCAO. Ultrastructural analysis of microvessels in nontransplant stroke rats revealed typical BBB pathology. At 5 days post-transplantation with hBMEPCs, stroke rats displayed widespread vascular repair in bilateral striatum and motor cortex, characterized by robust cell engraftment within capillaries. hBMEPC transplanted stroke rats exhibited near normal morphology of endothelial cells (ECs), pericytes, and astrocytes, without detectable perivascular edema. Near normal morphology of mitochondria was also detected in ECs and perivascular astrocytes from transplanted stroke rats. Equally notable, we observed numerous pinocytic vesicles within engrafted cells. Robust engraftment and intricate functionality of transplanted hBMEPCs likely abrogated stroke-altered vasculature. Preserving mitochondria and augmenting pinocytosis in cell-based therapeutics represent a new neurorestorative mechanism in BBB repair for stroke. Stem Cells 2017;35:1246-1258.
Collapse
Affiliation(s)
- Svitlana Garbuzova-Davis
- Center of Excellence for Aging & Brain Repair.,Department of Neurosurgery and Brain Repair.,Department of Molecular Pharmacology and Physiology.,Department of Pathology and Cell Biology, Morsani College of Medicine
| | - Edward Haller
- Department of Integrative Biology, University of South Florida, Tampa, Florida, USA
| | - Roger Lin
- Center of Excellence for Aging & Brain Repair
| | - Cesario V Borlongan
- Center of Excellence for Aging & Brain Repair.,Department of Neurosurgery and Brain Repair
| |
Collapse
|
30
|
Wang M, Li H, Si J, Dai J, Shi J, Wang X, Guo L, Shen G. Amniotic fluid-derived stem cells mixed with platelet rich plasma for restoration of rat alveolar bone defect. Acta Biochim Biophys Sin (Shanghai) 2017; 49:197-207. [PMID: 28104582 DOI: 10.1093/abbs/gmw133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Indexed: 12/12/2022] Open
Abstract
Stem cells isolated from the amniotic fluid have been shown as a promising candidate for cell therapy and tissue engineering. However, the experimental and preclinical applications of amniotic fluid-derived stem cells (AFSCs) in the very field of maxillofacial bone tissue engineering are still limited. In this study, rat AFSCs were successfully harvested and characterized in vitro. The rat AFSCs showed typical fibroblastoid morphology, stable proliferation activity and multi-differentiation potential. Flow-cytometry analysis demonstrated that these cells were positive for CD29, CD44, and CD90, while negative for hematopoietic markers such as CD34 and CD45. The regenerative performance of AFSCs-premixed with platelet rich plasma (PRP) gel in restoration of alveolar bone defect was further investigated using a modified rat maxillary alveolar defect model. Micro-computer tomography and histological examination showed a superior regenerative capacity of AFSCs-premixed with PRP gel at both 4 and 8 weeks after operation comparing with control groups. Moreover, the implanted AFSCs can survive in the defect site and directly participate in the bone tissue regeneration. Taken together, these results indicated the feasibility of an AFSCs-based alveolar bone tissue engineering strategy for alveolar defect restoration.
Collapse
Affiliation(s)
- Minjiao Wang
- Department of Oral and Craniomaxillofacial Science, Ninth People's Hospital College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Hongliang Li
- Department of Oral and Craniomaxillofacial Science, Ninth People's Hospital College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Jiawen Si
- Department of Oral and Craniomaxillofacial Science, Ninth People's Hospital College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Jiewen Dai
- Department of Oral and Craniomaxillofacial Science, Ninth People's Hospital College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Jun Shi
- Department of Oral and Craniomaxillofacial Science, Ninth People's Hospital College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Xudong Wang
- Department of Oral and Craniomaxillofacial Science, Ninth People's Hospital College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Lihe Guo
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guofang Shen
- Department of Oral and Craniomaxillofacial Science, Ninth People's Hospital College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| |
Collapse
|
31
|
Liang CC, Shaw SWS, Huang YH, Lin YH, Lee TH. Bladder Transplantation of Amniotic Fluid Stem Cell may Ameliorate Bladder Dysfunction After Focal Cerebral Ischemia in Rat. Stem Cells Transl Med 2017; 6:1227-1236. [PMID: 28186672 PMCID: PMC5442832 DOI: 10.1002/sctm.16-0212] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 07/29/2016] [Accepted: 10/31/2016] [Indexed: 12/20/2022] Open
Abstract
The objective is to investigate whether human amniotic fluid stem cells (hAFSCs) grafting into the bladder may influence bladder functional and molecular changes in an animal stroke model. Female rats were divided into three groups: sham, middle cerebral artery occlusion (MCAO) alone, and MCAO plus 1 × 106 hAFSCs transplanting into bladder wall. Bladder function was analyzed by cystometry at days 3 and 10 after MCAO. The expressions of bladder nerve growth factor (NGF), M2‐muscarinic, M3‐muscarinic, and P2X1 receptors were measured by immunohistochemistry and real‐time polymerase chain reaction. When compared with sham‐operated group, MCAO alone rats had significant increase in residual volume and decrease in voided volume and intercontraction interval; however, these bladder dysfunctions were improved following hAFSCs transplantation. The immunoreactivities of NGF, M3, and P2X1 significantly decreased at days 3 and 10, but M2 increased at day 3 after MCAO. Following hAFSCs transplantation, the immunoreactivities of NGF and P2X1 significantly increased at day 3, and M2 increased at day 10 after MCAO. The mRNAs of NGF, M2, and M3 significantly increased at day 3, but NGF and M2 decreased at day 10 after MCAO. Following hAFSCs transplantation, there was significant decrease in M2 mRNA at day 3 and increase in P2X1 mRNA at days 3 and 10 after MCAO. Bladder dysfunction caused by MCAO can be improved by hAFSCs transplanting into bladder which may be related to the expressions of bladder NGF, and muscarinic and P2X1 receptors. Stem Cells Translational Medicine2017;6:1227–1236
Collapse
Affiliation(s)
- Ching-Chung Liang
- Female Urology Section, Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan, Taiwan, Republic of China.,College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - S W Steven Shaw
- Division of Obstetrics, Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan, Taiwan, Republic of China.,College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China.,Prenatal Cell and Gene Therapy Group, Institute for Women's Health, University College London, London, United Kingdom
| | - Yung-Hsin Huang
- Female Urology Section, Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan, Taiwan, Republic of China
| | - Yi-Hao Lin
- Female Urology Section, Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan, Taiwan, Republic of China.,College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Tsong-Hai Lee
- Stroke Center and Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan, Taiwan, Republic of China.,College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China
| |
Collapse
|
32
|
Sokol J, Lippert T, Borlongan CV, Stuppia L. Translating amniotic fluid-derived stem cells for transplantation in stroke. Chin Neurosurg J 2016. [DOI: 10.1186/s41016-016-0055-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
33
|
Skardal A, Murphy SV, Crowell K, Mack D, Atala A, Soker S. A tunable hydrogel system for long-term release of cell-secreted cytokines and bioprinted in situ wound cell delivery. J Biomed Mater Res B Appl Biomater 2016; 105:1986-2000. [PMID: 27351939 DOI: 10.1002/jbm.b.33736] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 05/18/2016] [Accepted: 06/06/2016] [Indexed: 12/21/2022]
Abstract
For many cellular therapies being evaluated in preclinical and clinical trials, the mechanisms behind their therapeutic effects appear to be the secretion of growth factors and cytokines, also known as paracrine activity. Often, delivered cells are transient, and half-lives of the growth factors that they secrete are short, limiting their long-term effectiveness. The goal of this study was to optimize a hydrogel system capable of in situ cell delivery that could sequester and release growth factors secreted from those cells after the cells were no longer present. Here, we demonstrate the use of a fast photocross-linkable heparin-conjugated hyaluronic acid (HA-HP) hydrogel as a cell delivery vehicle for sustained growth factor release, which extends paracrine activity. The hydrogel could be modulated through cross-linking geometries and heparinization to support sustained release proteins and heparin-binding growth factors. To test the hydrogel in vivo, we used it to deliver amniotic fluid-derived stem (AFS) cells, which are known to secrete cytokines and growth factors, in full thickness skin wounds in a nu/nu murine model. Despite transience of the AFS cells in vivo, the HA-HP hydrogel with AFS cells improved wound closure and reepithelialization and increased vascularization and production of extracellular matrix in vivo. These results suggest that HA-HP hydrogel has the potential to prolong the paracrine activity of cells, thereby increasing their therapeutic effectiveness in wound healing. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 1986-2000, 2017.
Collapse
Affiliation(s)
- Aleksander Skardal
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina.,Department of Biomedical Engineering, Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Wake Forest Baptist Health, Medical Center Boulevard, Winston-Salem, North Carolina.,Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina
| | - Sean V Murphy
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina
| | - Kathryn Crowell
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina
| | - David Mack
- Department of Rehabilitation Medicine, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington.,Department of Bioengineering, University of Washington, Seattle, Washington
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina.,Department of Biomedical Engineering, Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Wake Forest Baptist Health, Medical Center Boulevard, Winston-Salem, North Carolina
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina.,Department of Biomedical Engineering, Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Wake Forest Baptist Health, Medical Center Boulevard, Winston-Salem, North Carolina.,Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina
| |
Collapse
|
34
|
Garbuzova-Davis S, Haller E, Tajiri N, Thomson A, Barretta J, Williams SN, Haim ED, Qin H, Frisina-Deyo A, Abraham JV, Sanberg PR, Van Loveren H, Borlongan CV. Blood-Spinal Cord Barrier Alterations in Subacute and Chronic Stages of a Rat Model of Focal Cerebral Ischemia. J Neuropathol Exp Neurol 2016; 75:673-88. [PMID: 27283328 DOI: 10.1093/jnen/nlw040] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We previously demonstrated blood-brain barrier impairment in remote contralateral brain areas in rats at 7 and 30 days after transient middle cerebral artery occlusion (tMCAO), indicating ischemic diaschisis. Here, we focused on effects of subacute and chronic focal cerebral ischemia on the blood-spinal cord barrier (BSCB). We observed BSCB damage on both sides of the cervical spinal cord in rats at 7 and 30 days post-tMCAO. Major BSCB ultrastructural changes in spinal cord gray and white matter included vacuolated endothelial cells containing autophagosomes, pericyte degeneration with enlarged mitochondria, astrocyte end-feet degeneration and perivascular edema; damaged motor neurons, swollen axons with unraveled myelin in ascending and descending tracts and astrogliosis were also observed. Evans Blue dye extravasation was maximal at 7 days. There was immunofluorescence evidence of reduction of microvascular expression of tight junction occludin, upregulation of Beclin-1 and LC3B immunoreactivities at 7 days and a reduction of the latter at 30 days post-ischemia. These novel pathological alterations on the cervical spinal cord microvasculature in rats after tMCAO suggest pervasive and long-lasting BSCB damage after focal cerebral ischemia, and that spinal cord ischemic diaschisis should be considered in the pathophysiology and therapeutic approaches in patients with ischemic cerebral infarction.
Collapse
Affiliation(s)
- Svitlana Garbuzova-Davis
- From the Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, NT, AT, JB, SNW, EDH, HQ, AF-D, JVA, PRS, CVB); Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS, HVL, CVB); Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D); Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS); Department of Integrative Biology, University of South Florida, Tampa, Florida (EH); Department of Psychiatry, Morsani College of Medicine, University of South Florida, Tampa, Florida (PRS).
| | - Edward Haller
- From the Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, NT, AT, JB, SNW, EDH, HQ, AF-D, JVA, PRS, CVB); Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS, HVL, CVB); Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D); Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS); Department of Integrative Biology, University of South Florida, Tampa, Florida (EH); Department of Psychiatry, Morsani College of Medicine, University of South Florida, Tampa, Florida (PRS)
| | - Naoki Tajiri
- From the Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, NT, AT, JB, SNW, EDH, HQ, AF-D, JVA, PRS, CVB); Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS, HVL, CVB); Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D); Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS); Department of Integrative Biology, University of South Florida, Tampa, Florida (EH); Department of Psychiatry, Morsani College of Medicine, University of South Florida, Tampa, Florida (PRS)
| | - Avery Thomson
- From the Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, NT, AT, JB, SNW, EDH, HQ, AF-D, JVA, PRS, CVB); Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS, HVL, CVB); Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D); Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS); Department of Integrative Biology, University of South Florida, Tampa, Florida (EH); Department of Psychiatry, Morsani College of Medicine, University of South Florida, Tampa, Florida (PRS)
| | - Jennifer Barretta
- From the Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, NT, AT, JB, SNW, EDH, HQ, AF-D, JVA, PRS, CVB); Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS, HVL, CVB); Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D); Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS); Department of Integrative Biology, University of South Florida, Tampa, Florida (EH); Department of Psychiatry, Morsani College of Medicine, University of South Florida, Tampa, Florida (PRS)
| | - Stephanie N Williams
- From the Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, NT, AT, JB, SNW, EDH, HQ, AF-D, JVA, PRS, CVB); Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS, HVL, CVB); Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D); Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS); Department of Integrative Biology, University of South Florida, Tampa, Florida (EH); Department of Psychiatry, Morsani College of Medicine, University of South Florida, Tampa, Florida (PRS)
| | - Eithan D Haim
- From the Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, NT, AT, JB, SNW, EDH, HQ, AF-D, JVA, PRS, CVB); Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS, HVL, CVB); Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D); Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS); Department of Integrative Biology, University of South Florida, Tampa, Florida (EH); Department of Psychiatry, Morsani College of Medicine, University of South Florida, Tampa, Florida (PRS)
| | - Hua Qin
- From the Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, NT, AT, JB, SNW, EDH, HQ, AF-D, JVA, PRS, CVB); Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS, HVL, CVB); Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D); Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS); Department of Integrative Biology, University of South Florida, Tampa, Florida (EH); Department of Psychiatry, Morsani College of Medicine, University of South Florida, Tampa, Florida (PRS)
| | - Aric Frisina-Deyo
- From the Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, NT, AT, JB, SNW, EDH, HQ, AF-D, JVA, PRS, CVB); Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS, HVL, CVB); Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D); Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS); Department of Integrative Biology, University of South Florida, Tampa, Florida (EH); Department of Psychiatry, Morsani College of Medicine, University of South Florida, Tampa, Florida (PRS)
| | - Jerry V Abraham
- From the Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, NT, AT, JB, SNW, EDH, HQ, AF-D, JVA, PRS, CVB); Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS, HVL, CVB); Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D); Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS); Department of Integrative Biology, University of South Florida, Tampa, Florida (EH); Department of Psychiatry, Morsani College of Medicine, University of South Florida, Tampa, Florida (PRS)
| | - Paul R Sanberg
- From the Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, NT, AT, JB, SNW, EDH, HQ, AF-D, JVA, PRS, CVB); Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS, HVL, CVB); Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D); Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS); Department of Integrative Biology, University of South Florida, Tampa, Florida (EH); Department of Psychiatry, Morsani College of Medicine, University of South Florida, Tampa, Florida (PRS)
| | - Harry Van Loveren
- From the Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, NT, AT, JB, SNW, EDH, HQ, AF-D, JVA, PRS, CVB); Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS, HVL, CVB); Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D); Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS); Department of Integrative Biology, University of South Florida, Tampa, Florida (EH); Department of Psychiatry, Morsani College of Medicine, University of South Florida, Tampa, Florida (PRS)
| | - Cesario V Borlongan
- From the Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, NT, AT, JB, SNW, EDH, HQ, AF-D, JVA, PRS, CVB); Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS, HVL, CVB); Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D); Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, Florida (SG-D, PRS); Department of Integrative Biology, University of South Florida, Tampa, Florida (EH); Department of Psychiatry, Morsani College of Medicine, University of South Florida, Tampa, Florida (PRS)
| |
Collapse
|
35
|
Gao L, Zhao M, Ye W, Huang J, Chu J, Yan S, Wang C, Zeng R. Inhibition of glycogen synthase kinase-3 (GSK3) promotes the neural differentiation of full-term amniotic fluid-derived stem cells towards neural progenitor cells. Tissue Cell 2016; 48:312-20. [PMID: 27346451 DOI: 10.1016/j.tice.2016.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 05/10/2016] [Accepted: 06/01/2016] [Indexed: 01/28/2023]
Abstract
The amniotic fluid has a heterogeneous population of cells. Some human amniotic fluid-derived stem (hAFS) cells have been shown to harbor the potential to differentiate into neural cells. However, the neural differentiation efficiency of hAFS cells remains low. In this study, we isolated CD117-positive hAFS cells from amniotic fluid and then examined the pluripotency of these cells through the formation of embryoid bodies (EBs). Additionally, we induced the neural differentiation of these cells using neuroectodermal medium. This study revealed that the GSK3-beta inhibitor SB216763 was able to stimulate the proliferation of CD117-positive hAFS cells without influencing their undifferentiated state. Moreover, SB216763 can efficiently promote the neural differentiation of CD117-positive hAFS cells towards neural progenitor cells in the presence of DMEM/F12 and N2 supplement. These findings provide an easy and low-cost method to maintain the proliferation of hAFS cells, as well as induce an efficacious generation of neural progenitor cells from hAFS cells. Such induction of the neural commitment of hAFS cells may provide an option for the treatment of neurodegenerative diseases by hAFS cells-based therapies.
Collapse
Affiliation(s)
- Liyang Gao
- Stem Cell Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| | - Mingyan Zhao
- Stem Cell Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Wei Ye
- Obstetrics and Gynecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jinzhi Huang
- Obstetrics and Gynecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiaqi Chu
- Stem Cell Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shouquan Yan
- Stem Cell Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Chaojun Wang
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Rong Zeng
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| |
Collapse
|
36
|
Amniotic Fluid Stem Cells: A Novel Source for Modeling of Human Genetic Diseases. Int J Mol Sci 2016; 17:ijms17040607. [PMID: 27110774 PMCID: PMC4849058 DOI: 10.3390/ijms17040607] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/01/2016] [Accepted: 04/11/2016] [Indexed: 12/15/2022] Open
Abstract
In recent years, great interest has been devoted to the use of Induced Pluripotent Stem cells (iPS) for modeling of human genetic diseases, due to the possibility of reprogramming somatic cells of affected patients into pluripotent cells, enabling differentiation into several cell types, and allowing investigations into the molecular mechanisms of the disease. However, the protocol of iPS generation still suffers from technical limitations, showing low efficiency, being expensive and time consuming. Amniotic Fluid Stem cells (AFS) represent a potential alternative novel source of stem cells for modeling of human genetic diseases. In fact, by means of prenatal diagnosis, a number of fetuses affected by chromosomal or Mendelian diseases can be identified, and the amniotic fluid collected for genetic testing can be used, after diagnosis, for the isolation, culture and differentiation of AFS cells. This can provide a useful stem cell model for the investigation of the molecular basis of the diagnosed disease without the necessity of producing iPS, since AFS cells show some features of pluripotency and are able to differentiate in cells derived from all three germ layers “in vitro”. In this article, we describe the potential benefits provided by using AFS cells in the modeling of human genetic diseases.
Collapse
|
37
|
Savitz SI, Parsha K. Enhancing Stroke Recovery with Cellular Therapies. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00060-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
38
|
Murphy SV, Hale A, Reid T, Olson J, Kidiyoor A, Tan J, Zhou Z, Jackson J, Atala A. Use of trimetasphere metallofullerene MRI contrast agent for the non-invasive longitudinal tracking of stem cells in the lung. Methods 2015; 99:99-111. [PMID: 26546729 DOI: 10.1016/j.ymeth.2015.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Revised: 10/18/2015] [Accepted: 11/03/2015] [Indexed: 12/31/2022] Open
Abstract
Magnetic Resonance Imaging (MRI) is a commonly used, non-invasive imaging technique that provides visualization of soft tissues with high spatial resolution. In both a research and clinical setting, the major challenge has been identifying a non-invasive and safe method for longitudinal tracking of delivered cells in vivo. The labeling and tracking of contrast agent labeled cells using MRI has the potential to fulfill this need. Contrast agents are often used to enhance the image contrast between the tissue of interest and surrounding tissues with MRI. The most commonly used MRI contrast agents contain Gd(III) ions. However, Gd(III) ions are highly toxic in their ionic form, as they tend to accumulate in the liver, spleen, kidney and bones and block calcium channels. Endohedral metallofullerenes such as trimetallic nitride endohedral metallofullerenes (Trimetasphere®) are one unique class of fullerene molecules where a Gd3N cluster is encapsulated inside a C80 carbon cage referred to as Gd3N@C80. These endohedral metallofullerenes have several advantages over small chelated Gd(III) complexes such as increased stability of the Gd(III) ion, minimal toxic effects, high solubility in water and high proton relativity. In this study, we describe the evaluation of gadolinium-based Trimetasphere® positive contrast agent for the in vitro labeling and in vivo tracking of human amniotic fluid-derived stem cells within lung tissue. In addition, we conducted a 'proof-of-concept' experiment demonstrating that this methodology can be used to track the homing of stem cells to injured lung tissue and provide longitudinal analysis of cell localization over an extended time course.
Collapse
Affiliation(s)
- Sean V Murphy
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| | - Austin Hale
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| | - Tanya Reid
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| | - John Olson
- Center for Biomolecular Imaging, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA.
| | - Amritha Kidiyoor
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| | - Josh Tan
- Center for Biomolecular Imaging, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA.
| | - Zhiguo Zhou
- Luna nanoWorks Division, Luna Innovations, Incorporated, Danville, VA 24541, USA.
| | - John Jackson
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| |
Collapse
|
39
|
de la Peña I, Sanberg PR, Acosta S, Lin SZ, Borlongan CV. Umbilical cord blood cell and granulocyte-colony stimulating factor: combination therapy for traumatic brain injury. Regen Med 2015; 9:409-12. [PMID: 25159056 DOI: 10.2217/rme.14.32] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Ike de la Peña
- Department of Neurosurgery & Brain Repair, Center of Excellence for Aging & Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | | | | | | | | |
Collapse
|
40
|
Jendelová P, Kubinová Š, Sandvig I, Erceg S, Sandvig A, Syková E. Current developments in cell- and biomaterial-based approaches for stroke repair. Expert Opin Biol Ther 2015; 16:43-56. [DOI: 10.1517/14712598.2016.1094457] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
41
|
Garbuzova-Davis S, Haller E, Williams SN, Haim ED, Tajiri N, Hernandez-Ontiveros DG, Frisina-Deyo A, Boffeli SM, Sanberg PR, Borlongan CV. Compromised blood-brain barrier competence in remote brain areas in ischemic stroke rats at the chronic stage. J Comp Neurol 2015; 522:3120-37. [PMID: 24610730 DOI: 10.1002/cne.23582] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 01/28/2014] [Accepted: 03/05/2014] [Indexed: 12/14/2022]
Abstract
Stroke is a life-threatening disease leading to long-term disability in stroke survivors. Cerebral functional insufficiency in chronic stroke might be due to pathological changes in brain areas remote from the initial ischemic lesion, i.e., diaschisis. Previously, we showed that the damaged blood-brain barrier (BBB) was involved in subacute diaschisis. The present study investigated BBB competence in chronic diaschisis by using a transient middle cerebral artery occlusion (tMCAO) rat model. Our results demonstrated significant BBB damage mostly in the ipsilateral striatum and motor cortex in rats at 30 days after tMCAO. The BBB alterations were also determined in the contralateral hemisphere via ultrastructural and immunohistochemical analyses. Major BBB pathological changes in contralateral remote striatum and motor cortex areas included 1) vacuolated endothelial cells containing large autophagosomes, 2) degenerated pericytes displaying mitochondria with cristae disruption, 3) degenerated astrocytes and perivascular edema, 4) Evans blue extravasation, and 5) appearance of parenchymal astrogliosis. Discrete analyses of striatal and motor cortex areas revealed significantly higher autophagosome accumulation in capillaries of ventral striatum and astrogliosis in dorsal striatum in both cerebral hemispheres. These widespread microvascular alterations in ipsilateral and contralateral brain hemispheres suggest persistent and/or continued BBB damage in chronic ischemia. The pathological changes in remote brain areas likely indicate chronic ischemic diaschisis, which should be considered in the development of treatment strategies for stroke.
Collapse
Affiliation(s)
- Svitlana Garbuzova-Davis
- Center of Excellence for Aging and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612; Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612; Department of Molecular Pharmacology and Physiology, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612; Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, Florida, 33612
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Stem cell transplantation (SCT) is an established first-line or adjunctive therapy for a variety of neonatal and adult diseases. New evidence in preclinical models as well as a few human studies show the potential utility of SCT in neuroprotection and in the modulation of inflammatory injury in at risk-neonates. This review briefly summarizes current understanding of human stem cell biology during ontogeny and present recent evidence supporting SCT as a viable approach for postinsult neonatal injury.
Collapse
Affiliation(s)
- Momoko Yoshimoto
- Assistant Research Professor, Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, 1044W Walnut Street R4-W116, Indianapolis, IN 46202, Tel: 317-278-0598
| | - Joyce M Koenig
- Pediatrics, E Doisy Research Center, Saint Louis University School of Medicine, 1100 South Grand Boulevard, St Louis, MO 63104, USA; Molecular Microbiology & Immunology, E Doisy Research Center, Saint Louis University School of Medicine, 1100 South Grand Boulevard, St Louis, MO 63106, USA.
| |
Collapse
|
43
|
Elias M, Hoover J, Nguyen H, Reyes S, Lawton C, Borlongan CV. Stroke therapy: the potential of amniotic fluid-derived stem cells. FUTURE NEUROLOGY 2015; 10:321-326. [PMID: 26401122 DOI: 10.2217/fnl.15.19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Maya Elias
- Department of Neurosurgery & Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, FL 33612, USA
| | - Jaclyn Hoover
- Department of Neurosurgery & Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, FL 33612, USA
| | - Hung Nguyen
- Department of Neurosurgery & Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, FL 33612, USA
| | - Stephanny Reyes
- Department of Neurosurgery & Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, FL 33612, USA
| | - Christopher Lawton
- Department of Neurosurgery & Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, FL 33612, USA
| | - Cesar V Borlongan
- Department of Neurosurgery & Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, FL 33612, USA ; Center of Excellence for Aging & Brain Repair, Department of Neurosurgery & Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| |
Collapse
|
44
|
Praet J, Santermans E, Daans J, Le Blon D, Hoornaert C, Goossens H, Hens N, Van Der Linden A, Berneman Z, Ponsaerts P. Early Inflammatory Responses following Cell Grafting in the CNS Trigger Activation of the Subventricular Zone: A Proposed Model of Sequential Cellular Events. Cell Transplant 2015; 24:1481-92. [DOI: 10.3727/096368914x682800] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
While multiple rodent preclinical studies, and to a lesser extent human clinical trials, claim the feasibility, safety, and potential clinical benefit of cell grafting in the central nervous system (CNS), currently only little convincing knowledge exists regarding the actual fate of the grafted cells and their effect on the surrounding environment (or vice versa). Our preceding studies already indicated that only a minor fraction of the initially grafted cell population survives the grafting process, while the surviving cell population becomes invaded by highly activated microglia/macrophages and surrounded by reactive astrogliosis. In the current study, we further elaborate on early cellular and inflammatory events following syngeneic grafting of eGFP mouse embryonic fibroblasts (mEFs) in the CNS of immunocompetent mice. Based on obtained quantitative histological data, we here propose a detailed mathematically derived working model that sequentially comprises hypoxia-induced apoptosis of grafted mEFs, neutrophil invasion, neoangiogenesis, microglia/macrophage recruitment, astrogliosis, and eventually survival of a limited number of grafted mEFs. Simultaneously, we observed that the cellular events following mEF grafting activates the subventricular zone neural stem and progenitor cell compartment. This proposed model therefore further contributes to our understanding of cell graft-induced cellular responses and will eventually allow for successful manipulation of this intervention.
Collapse
Affiliation(s)
- Jelle Praet
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
- BioImaging Laboratory, University of Antwerp, Wilrijk, Belgium
| | - Eva Santermans
- Center for Statistics, I-Biostat, Hasselt University, Diepenbeek, Belgium
| | - Jasmijn Daans
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Debbie Le Blon
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Chloé Hoornaert
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Herman Goossens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Niel Hens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
- Center for Statistics, I-Biostat, Hasselt University, Diepenbeek, Belgium
- Centre for Health Economic Research and Modeling Infectious Diseases (Chermid), University of Antwerp, Wilrijk, Belgium
| | | | - Zwi Berneman
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Peter Ponsaerts
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
45
|
Acosta SA, Tajiri N, Hoover J, Kaneko Y, Borlongan CV. Intravenous Bone Marrow Stem Cell Grafts Preferentially Migrate to Spleen and Abrogate Chronic Inflammation in Stroke. Stroke 2015. [PMID: 26219646 PMCID: PMC4542567 DOI: 10.1161/strokeaha.115.009854] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND PURPOSE Adult stem cell therapy is an experimental stroke treatment. Here, we assessed homing and anti-inflammatory effects of bone marrow stromal cells (hBMSCs) in chronic stroke. METHODS At 60 days post stroke, adult Sprague-Dawley rats received intravenous hBMSCs (4×10(6) labeled or nonlabeled cells) or vehicle (saline). A sham surgery group served as additional control. In vivo imaging was conducted between 1 hour and 11 days post transplantation, followed by histological examination. RESULTS Labeled hBMSCs migrated to spleen which emitted significantly higher fluorescent signal across all time points, especially during the first hour, and were modestly detected in the head region at the 12 hours and 11 days, compared with nonlabeled hBMSCs and vehicle-infused stroke animals, or sham (P<0.05). At 11 days post transplantation, ex vivo imaging confirmed preferential hBMSC migration to the spleen over the brain. Hematoxylin and eosin staining revealed significant 15% and 30% reductions in striatal infarct and peri-infarct area, and a trend of rescue against neuronal loss in the hippocampus. Unbiased stereology showed significant 75% and 60% decrements in major histocompatibility complex II-activated inflammatory cells in gray and white matter, and a 43% diminution in tumor necrosis factor-α cell density in the spleen of transplanted stroke animals compared with vehicle-infused stroke animals (P<0.05). Human antigen immunostaining revealed 0.03% hBMSCs survived in spleen and only 0.0007% in brain. MSC migration to spleen, but not brain, inversely correlated with reduced infarct, peri-infarct, and inflammation. CONCLUSIONS hBMSC transplantation is therapeutic in chronic stroke possibly by abrogating the inflammation-plagued secondary cell death.
Collapse
Affiliation(s)
- Sandra A Acosta
- From the Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa
| | - Naoki Tajiri
- From the Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa
| | - Jaclyn Hoover
- From the Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa
| | - Yuji Kaneko
- From the Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa
| | - Cesar V Borlongan
- From the Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa.
| |
Collapse
|
46
|
Abstract
The field of stem cell therapy has emerged as a promising research area for brain repair. Optimizing the safety and efficacy of the therapy for clinical trials will require revisiting transplantation protocols. The cell delivery route stands as a key translational item that warrants careful consideration in facilitating the success of stem cell therapy in the clinic. Intracerebral administration, compared to peripheral route, requires an invasive procedure to directly implant stem cells into injured brain. Although invasive, intracerebral transplantation circumvents the prohibitive blood brain barrier in allowing grafted cells when delivered peripherally to penetrate the brain and reach the discreet damaged brain tissues. This review will highlight milestone discoveries in cell therapy for neurological disorders, with emphasis on intracerebral transplantation in relevant animal models and provide insights necessary to optimize the safety and efficacy of cell therapy for the treatment of Parkinson's disease, Huntington's disease, stroke and traumatic brain injury.
Collapse
Affiliation(s)
- Stephanny Reyes
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, MDC 78, 12901 Bruce B. Downs Blvd, Tampa, FL 33612, USA
| | | | | |
Collapse
|
47
|
Amniotic Fluid-Derived Stem Cells (AFSC) and Their Application in Cell Therapy and Tissue Engineering. RAZAVI INTERNATIONAL JOURNAL OF MEDICINE 2015. [DOI: 10.5812/rijm.20135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
48
|
Detante O, Jaillard A, Moisan A, Barbieux M, Favre I, Garambois K, Hommel M, Remy C. Biotherapies in stroke. Rev Neurol (Paris) 2014; 170:779-98. [DOI: 10.1016/j.neurol.2014.10.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 09/29/2014] [Accepted: 10/08/2014] [Indexed: 12/31/2022]
|
49
|
Wei PC, Chao A, Peng HH, Chao AS, Chang YL, Chang SD, Wang HS, Chang YJ, Tsai MS, Sieber M, Chen HC, Chen SJ, Lee YS, Hwang SM, Wang TH. SOX9 as a Predictor for Neurogenesis Potentiality of Amniotic Fluid Stem Cells. Stem Cells Transl Med 2014; 3:1138-47. [PMID: 25154783 DOI: 10.5966/sctm.2014-0019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Preclinical studies of amniotic fluid-derived cell therapy have been successful in the research of neurodegenerative diseases, peripheral nerve injury, spinal cord injury, and brain ischemia. Transplantation of human amniotic fluid stem cells (AFSCs) into rat brain ventricles has shown improvement in symptoms of Parkinson's disease and also highlighted the minimal immune rejection risk of AFSCs, even between species. Although AFSCs appeared to be a promising resource for cell-based regenerative therapy, AFSCs contain a heterogeneous pool of distinct cell types, rendering each preparation of AFSCs unique. Identification of predictive markers for neuron-prone AFSCs is necessary before such stem cell-based therapeutics can become a reality. In an attempt to identify markers of AFSCs to predict their ability for neurogenesis, we performed a two-phase study. In the discovery phase of 23 AFSCs, we tested ZNF521/Zfp521, OCT6, SOX1, SOX2, SOX3, and SOX9 as predictive markers of AFSCs for neural differentiation. In the validation phase, the efficacy of these predictive markers was tested in independent sets of 18 AFSCs and 14 dental pulp stem cells (DPSCs). We found that high expression of SOX9 in AFSCs is associated with good neurogenetic ability, and these positive correlations were confirmed in independent sets of AFSCs and DPSCs. Furthermore, knockdown of SOX9 in AFSCs inhibited their neuronal differentiation. In conclusion, the discovery of SOX9 as a predictive marker for neuron-prone AFSCs could expedite the selection of useful clones for regenerative medicine, in particular, in neurological diseases and injuries.
Collapse
Affiliation(s)
- Pei-Cih Wei
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Angel Chao
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Hsiu-Huei Peng
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - An-Shine Chao
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Yao-Lung Chang
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Shuenn-Dyh Chang
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Hsin-Shih Wang
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Yu-Jen Chang
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Ming-Song Tsai
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Martin Sieber
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Hua-Chien Chen
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Shu-Jen Chen
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Yun-Shien Lee
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Shiaw-Min Hwang
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Tzu-Hao Wang
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| |
Collapse
|
50
|
Tajiri N, Acosta S, Portillo-Gonzales GS, Aguirre D, Reyes S, Lozano D, Pabon M, Dela Peña I, Ji X, Yasuhara T, Date I, Solomita MA, Antonucci I, Stuppia L, Kaneko Y, Borlongan CV. Therapeutic outcomes of transplantation of amniotic fluid-derived stem cells in experimental ischemic stroke. Front Cell Neurosci 2014; 8:227. [PMID: 25165432 PMCID: PMC4131212 DOI: 10.3389/fncel.2014.00227] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 07/23/2014] [Indexed: 01/29/2023] Open
Abstract
Accumulating preclinical evidence suggests the use of amnion as a source of stem cells for investigations of basic science concepts related to developmental cell biology, but also for stem cells’ therapeutic applications in treating human disorders. We previously reported isolation of viable rat amniotic fluid-derived stem (AFS) cells. Subsequently, we recently reported the therapeutic benefits of intravenous transplantation of AFS cells in a rodent model of ischemic stroke. Parallel lines of investigations have provided safety and efficacy of stem cell therapy for treating stroke and other neurological disorders. This review article highlights the need for investigations of mechanisms underlying AFS cells’ therapeutic benefits and discusses lab-to-clinic translational gating items in an effort to optimize the clinical application of the cell transplantation for stroke.
Collapse
Affiliation(s)
- Naoki Tajiri
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Sandra Acosta
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Gabriel S Portillo-Gonzales
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Daniela Aguirre
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Stephanny Reyes
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Diego Lozano
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Mibel Pabon
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Ike Dela Peña
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University Beijing, China
| | - Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences Okayama, Japan
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences Okayama, Japan
| | - Marianna A Solomita
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Ivana Antonucci
- Laboratory of Molecular Genetics, DISPUTer, School of Medicine and Health Sciences, "G. d 'Annunzio" University Chieti-Pescara, Italy
| | - Liborio Stuppia
- Laboratory of Molecular Genetics, DISPUTer, School of Medicine and Health Sciences, "G. d 'Annunzio" University Chieti-Pescara, Italy
| | - Yuji Kaneko
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine Tampa, FL, USA
| |
Collapse
|