1
|
Kumar R, Mahajan S, Gupta U, Madan J, Godugu C, Guru SK, Singh PK, Parvatikar P, Maji I. Stem cell therapy as a novel concept to combat CNS disorders. TARGETED THERAPY FOR THE CENTRAL NERVOUS SYSTEM 2025:175-206. [DOI: 10.1016/b978-0-443-23841-3.00009-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
2
|
Calderazzo S, Covert M, Alba DD, Bowley BE, Pessina MA, Rosene DL, Buller B, Medalla M, Moore TL. Neural recovery after cortical injury: Effects of MSC derived extracellular vesicles on motor circuit remodeling in rhesus monkeys. IBRO Neurosci Rep 2022; 13:243-254. [PMID: 36590089 PMCID: PMC9795302 DOI: 10.1016/j.ibneur.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 07/01/2022] [Accepted: 08/07/2022] [Indexed: 01/04/2023] Open
Abstract
Reorganization of motor circuits in the cortex and corticospinal tract are thought to underlie functional recovery after cortical injury, but the mechanisms of neural plasticity that could be therapeutic targets remain unclear. Recent work from our group have shown that systemic treatment with mesenchymal stem cell derived (MSCd) extracellular vesicles (EVs) administered after cortical damage to the primary motor cortex (M1) of rhesus monkeys resulted in a robust recovery of fine motor function and reduced chronic inflammation. Here, we used immunohistochemistry for cfos, an activity-dependent intermediate early gene, to label task-related neurons in the surviving primary motor and premotor cortices, and markers of axonal and synaptic plasticity in the spinal cord. Compared to vehicle, EV treatment was associated with a greater density of cfos+ pyramidal neurons in the deep layers of M1, greater density of cfos+ inhibitory interneurons in premotor areas, and lower density of synapses on MAP2+ lower motor neurons in the cervical spinal cord. These data suggest that the anti-inflammatory effects of EVs may reduce injury-related upper motor neuron damage and hyperexcitability, as well as aberrant compensatory re-organization in the cervical spinal cord to improve motor function.
Collapse
Key Words
- CB, Calbindin
- CR, Calretinin
- CSC, Cervical Spinal Cord
- Circuit Remodeling
- Cortical Injury
- DH, Dorsal Horn
- EVs, Extracellular Vesicles
- Extracellular Vesicles
- Ischemia
- LCST, Lateral Corticospinal Tract
- M1, Primary Motor Cortex
- MAP2, Microtubule Associated Protein 2
- MSCd, Mesenchymal Stem Cell derived
- Motor Cortex
- NHP, Non-Human Primate
- PV, Parvalbumin
- Plasticity
- ROS, Reactive Oxygen Species
- SYN, Synaptophysin
- Stem Cell-Based Treatments
- VH, Ventral Horn
- dPMC, dorsal Premotor Cortex
- miRNA, Micro RNA
- periM1, Perilesional Primary Motor Cortex
Collapse
Affiliation(s)
| | | | | | | | | | - Douglas L. Rosene
- Anatomy and Neurobiology Dept, BUSM, USA
- Center for Systems Neuroscience, BU, USA
| | | | - Maria Medalla
- Anatomy and Neurobiology Dept, BUSM, USA
- Center for Systems Neuroscience, BU, USA
| | - Tara L. Moore
- Anatomy and Neurobiology Dept, BUSM, USA
- Center for Systems Neuroscience, BU, USA
| |
Collapse
|
3
|
Venugopal C, Shobha K, Rai KS, Dhanushkodi A. Neurogenic and cognitive enhancing effects of human dental pulp stem cells and its secretome in animal model of hippocampal neurodegeneration. Brain Res Bull 2022; 180:46-58. [PMID: 34979238 DOI: 10.1016/j.brainresbull.2021.12.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 11/11/2021] [Accepted: 12/28/2021] [Indexed: 12/26/2022]
Abstract
Progressive hippocampal neuronal losses, neuroinflammation, declined neurogenesis and impaired hippocampal functions are pathological features of Alzheimer's disease and temporal lobe epilepsy (TLE). Halting neuroinflammation and progressive neurodegeneration in the hippocampus is a major challenge in treating such disease conditions which, if unsuccessful would lead to learning/memory dysfunction and co-morbidities like anxiety/depression. Mesenchymal stem cells (MSCs) therapy provides hope for treating neurodegenerative diseases by either replacing lost neurons by transplantation of MSCs which might differentiate into appropriate neuronal phenotypes or by stimulating the resident neural stem cells for proliferation/differentiation. In this current study, we demonstrate that the intrahippocampal transplantation of ectoderm originated dental pulp stem cells (DPSCs) or intrahippocampal injection of DPSCs condition medium (DPSCs-CM) in a mouse model of hippocampal neurodegeneration could efficiently prevent neurodegeneration, neuroinflammation, enhance hippocampal neurogenesis and spatial learning and memory functions much superior to commonly used bone marrow mesenchymal stem cells (BM-MSCs) or its secretome. Probing the possible mechanisms of neuroprotection revealed that DPSCs/DPSCs-CM treatment upregulated an array of hosts' endogenous neural survival factors expression, reduced pro-apoptotic caspase activity and upregulated the anti-apoptotic factors BCL-2 and phosphorylated PI3K prominently than BM-MSCs/BM-MSCs-CM, suggesting that among MSCs, neural crest originated DPSCs might be a better adult stem cell candidate for treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Chaitra Venugopal
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - K Shobha
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Kiranmai S Rai
- Dept. of Physiology, Melaka Manipal Medical College, Manipal Academy of Higher, Education, Manipal, Karnataka, India
| | - Anandh Dhanushkodi
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
4
|
Pischiutta F, Caruso E, Lugo A, Cavaleiro H, Stocchetti N, Citerio G, Salgado A, Gallus S, Zanier ER. Systematic review and meta-analysis of preclinical studies testing mesenchymal stromal cells for traumatic brain injury. NPJ Regen Med 2021; 6:71. [PMID: 34716332 PMCID: PMC8556393 DOI: 10.1038/s41536-021-00182-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/30/2021] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are widely used in preclinical models of traumatic brain injury (TBI). Results are promising in terms of neurological improvement but are hampered by wide variability in treatment responses. We made a systematic review and meta-analysis: (1) to assess the quality of evidence for MSC treatment in TBI rodent models; (2) to determine the effect size of MSCs on sensorimotor function, cognitive function, and anatomical damage; (3) to identify MSC-related and protocol-related variables associated with greater efficacy; (4) to understand whether MSC manipulations boost therapeutic efficacy. The meta-analysis included 80 studies. After TBI, MSCs improved sensorimotor and cognitive deficits and reduced anatomical damage. Stratified meta-analysis on sensorimotor outcome showed similar efficacy for different MSC sources and for syngeneic or xenogenic transplants. Efficacy was greater when MSCs were delivered in the first-week post-injury, and when implanted directly into the lesion cavity. The greatest effect size was for cells embedded in matrices or for MSC-derivatives. MSC therapy is effective in preclinical TBI models, improving sensorimotor, cognitive, and anatomical outcomes, with large effect sizes. These findings support clinical studies in TBI.
Collapse
Affiliation(s)
- Francesca Pischiutta
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Enrico Caruso
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.,Neuroscience Intensive Care Unit, Department of Anesthesia and Critical Care, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alessandra Lugo
- Laboratory of Lifestyle Epidemiology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Helena Cavaleiro
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.,Stemmatters, Biotechnology and Regenerative Medicine, Guimarães, Portugal
| | - Nino Stocchetti
- Neuroscience Intensive Care Unit, Department of Anesthesia and Critical Care, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplants, University of Milan, Milan, Italy
| | - Giuseppe Citerio
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - António Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Silvano Gallus
- Laboratory of Lifestyle Epidemiology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Elisa R Zanier
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| |
Collapse
|
5
|
Bonilla C, Zurita M. Cell-Based Therapies for Traumatic Brain Injury: Therapeutic Treatments and Clinical Trials. Biomedicines 2021; 9:biomedicines9060669. [PMID: 34200905 PMCID: PMC8230536 DOI: 10.3390/biomedicines9060669] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury (TBI) represents physical damage to the brain tissue that induces transitory or permanent neurological disabilities. TBI contributes to 50% of all trauma deaths, with many enduring long-term consequences and significant medical and rehabilitation costs. There is currently no therapy to reverse the effects associated with TBI. An increasing amount of research has been undertaken regarding the use of different stem cells (SCs) to treat the consequences of brain damage. Neural stem cells (NSCs) (adult and embryonic) and mesenchymal stromal cells (MSCs) have shown efficacy in pre-clinical models of TBI and in their introduction to clinical research. The purpose of this review is to provide an overview of TBI and the state of clinical trials aimed at evaluating the use of stem cell-based therapies in TBI. The primary aim of these studies is to investigate the safety and efficacy of the use of SCs to treat this disease. Although an increasing number of studies are being carried out, few results are currently available. In addition, we present our research regarding the use of cell therapy in TBI. There is still a significant lack of understanding regarding the cell therapy mechanisms for the treatment of TBI. Thus, future studies are needed to evaluate the feasibility of the transplantation of SCs in TBI.
Collapse
Affiliation(s)
- Celia Bonilla
- Cell Therapy Unit, Puerta de Hierro Hospital, 28222 Majadahonda, Madrid, Spain
- Correspondence: ; Tel.: +34-91-191-7879
| | - Mercedes Zurita
- Cell Therapy Unit Responsable, Puerta de Hierro Hospital, 28222 Majadahonda, Madrid, Spain;
| |
Collapse
|
6
|
Das M, Mayilsamy K, Mohapatra SS, Mohapatra S. Mesenchymal stem cell therapy for the treatment of traumatic brain injury: progress and prospects. Rev Neurosci 2020; 30:839-855. [PMID: 31203262 DOI: 10.1515/revneuro-2019-0002] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/05/2019] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury (TBI) is a major cause of injury-related mortality and morbidity in the USA and around the world. The survivors may suffer from cognitive and memory deficits, vision and hearing loss, movement disorders, and different psychological problems. The primary insult causes neuronal damage and activates astrocytes and microglia which evokes immune responses causing further damage to the brain. Clinical trials of drugs to recover the neuronal loss are not very successful. Regenerative approaches for TBI using mesenchymal stem cells (MSCs) seem promising. Results of preclinical research have shown that transplantation of MSCs reduced secondary neurodegeneration and neuroinflammation, promoted neurogenesis and angiogenesis, and improved functional outcome in the experimental animals. The functional improvement is not necessarily related to cell engraftment; rather, immunomodulation by molecular factors secreted by MSCs is responsible for the beneficial effects of this therapy. However, MSC therapy has a few drawbacks including tumor formation, which can be avoided by the use of MSC-derived exosomes. This review has focused on the research works published in the field of regenerative therapy using MSCs after TBI and its future direction.
Collapse
Affiliation(s)
- Mahasweta Das
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA.,Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Karthick Mayilsamy
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA.,Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Shyam S Mohapatra
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA.,Department of Internal Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Subhra Mohapatra
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA.,Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| |
Collapse
|
7
|
Zhang Y, Zhang Y, Chopp M, Zhang ZG, Mahmood A, Xiong Y. Mesenchymal Stem Cell-Derived Exosomes Improve Functional Recovery in Rats After Traumatic Brain Injury: A Dose-Response and Therapeutic Window Study. Neurorehabil Neural Repair 2020; 34:616-626. [PMID: 32462980 DOI: 10.1177/1545968320926164] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background. Mesenchymal stem cell (MSC)-derived exosomes play a critical role in regenerative medicine. Objective. To determine the dose- and time-dependent efficacy of exosomes for treatment of traumatic brain injury (TBI). Methods. Male rats were subjected to a unilateral moderate cortical contusion. In the dose-response study, animals received a single intravenous injection of exosomes (50, 100, 200 µg per rat) or vehicle, with treatment initiated at 1 day after injury. In the therapeutic window study, animals received a single intravenous injection of 100 µg exosomes or vehicle starting at 1, 4, or 7 days after injury. Neurological functional tests were performed weekly after TBI for 5 weeks. Spatial learning was measured on days 31 to 35 after TBI using the Morris water maze test. Results. Compared with the vehicle, regardless of the dose and delay in treatment, exosome treatment significantly improved sensorimotor and cognitive function, reduced hippocampal neuronal cell loss, promoted angiogenesis and neurogenesis, and reduced neuroinflammation. Exosome treatment at 100 µg per rat exhibited a significant therapeutic effect compared with the 50- or 200-µg exosome groups. The time-dependent exosome treatment data demonstrated that exosome treatment starting at 1 day post-TBI provided a significantly greater improvement in functional and histological outcomes than exosome treatments at the other 2 delayed treatments. Conclusions. These results indicate that exosomes have a wide range of effective doses for treatment of TBI with a therapeutic window of at least 7 days postinjury. Exosomes may provide a novel therapeutic intervention in TBI.
Collapse
Affiliation(s)
| | - Yi Zhang
- Henry Ford Hospital, Detroit, MI, USA
| | - Michael Chopp
- Henry Ford Hospital, Detroit, MI, USA.,Oakland University, Rochester, MI, USA
| | | | | | - Ye Xiong
- Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
8
|
Ruppert KA, Prabhakara KS, Toledano-Furman NE, Udtha S, Arceneaux AQ, Park H, Dao A, Cox CS, Olson SD. Human adipose-derived mesenchymal stem cells for acute and sub-acute TBI. PLoS One 2020; 15:e0233263. [PMID: 32453741 PMCID: PMC7250455 DOI: 10.1371/journal.pone.0233263] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 04/20/2020] [Indexed: 02/07/2023] Open
Abstract
In the U.S., approximately 1.7 million people suffer traumatic brain injury each year, with many enduring long-term consequences and significant medical and rehabilitation costs. The primary injury causes physical damage to neurons, glia, fiber tracts and microvasculature, which is then followed by secondary injury, consisting of pathophysiological mechanisms including an immune response, inflammation, edema, excitotoxicity, oxidative damage, and cell death. Most attempts at intervention focus on protection, repair or regeneration, with regenerative medicine becoming an intensively studied area over the past decade. The use of stem cells has been studied in many disease and injury models, using stem cells from a variety of sources and applications. In this study, human adipose-derived mesenchymal stromal cells (MSCs) were administered at early (3 days) and delayed (14 days) time points after controlled cortical impact (CCI) injury in rats. Animals were routinely assessed for neurological and vestibulomotor deficits, and at 32 days post-injury, brain tissue was processed by flow cytometry and immunohistochemistry to analyze neuroinflammation. Treatment with HB-adMSC at either 3d or 14d after injury resulted in significant improvements in neurocognitive outcome and a change in neuroinflammation one month after injury.
Collapse
Affiliation(s)
- Katherine A. Ruppert
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Karthik S. Prabhakara
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Naama E. Toledano-Furman
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Sanjna Udtha
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Austin Q. Arceneaux
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | | | - An Dao
- Hope Biosciences, Sugarland, TX, United States of America
| | - Charles S. Cox
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Scott D. Olson
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States of America
- * E-mail:
| |
Collapse
|
9
|
Moore TL, Bowley BGE, Pessina MA, Calderazzo SM, Medalla M, Go V, Zhang ZG, Chopp M, Finklestein S, Harbaugh AG, Rosene DL, Buller B. Mesenchymal derived exosomes enhance recovery of motor function in a monkey model of cortical injury. Restor Neurol Neurosci 2020; 37:347-362. [PMID: 31282441 DOI: 10.3233/rnn-190910] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Exosomes from mesenchymal stromal cells (MSCs) are endosome-derived vesicles that have been shown to enhance functional recovery in rodent models of stroke. OBJECTIVE Building on these findings, we tested exosomes as a treatment in monkeys with cortical injury. METHODS After being trained on a task of fine motor function of the hand, monkeys received a cortical injury to the hand representation in primary motor cortex. Twenty-four hours later and again 14 days after injury, monkeys received exosomes or vehicle control. Recovery of motor function was followed for 12 weeks. RESULTS Compared to monkeys that received vehicle, exosome treated monkeys returned to pre-operative grasp patterns and latency to retrieve a food reward in the first three-five weeks of recovery. CONCLUSIONS These results provide evidence that in monkeys exosomes delivered after cortical injury enhance recovery of motor function.
Collapse
Affiliation(s)
- T L Moore
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA
| | - B G E Bowley
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA
| | - M A Pessina
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA
| | - S M Calderazzo
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA
| | - M Medalla
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA
| | - V Go
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Z G Zhang
- Department of Neurology, Henry Ford Health Systems, Detroit, MI, USA
| | - M Chopp
- Department of Neurology, Henry Ford Health Systems, Detroit, MI, USA
| | - S Finklestein
- Stemetix, Inc. Needham, MA, USA.,Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - A G Harbaugh
- Department Mathematics & Statistics, Boston University, Boston, MA, USA
| | - D L Rosene
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA
| | - B Buller
- Department of Neurology, Henry Ford Health Systems, Detroit, MI, USA
| |
Collapse
|
10
|
Schepici G, Silvestro S, Bramanti P, Mazzon E. Traumatic Brain Injury and Stem Cells: An Overview of Clinical Trials, the Current Treatments and Future Therapeutic Approaches. ACTA ACUST UNITED AC 2020; 56:medicina56030137. [PMID: 32204311 PMCID: PMC7143935 DOI: 10.3390/medicina56030137] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/04/2020] [Accepted: 03/15/2020] [Indexed: 02/07/2023]
Abstract
Traumatic brain injury represents physical damage to the brain tissue that induces transitory or permanent neurological disabilities. The traumatic injury activates an important inflammatory response, followed by a cascade of events that lead to neuronal loss and further brain damage. Maintaining proper ventilation, a normal level of oxygenation, and adequate blood pressure are the main therapeutic strategies performed after injury. Surgery is often necessary for patients with more serious injuries. However, to date, there are no therapies that completely resolve the brain damage suffered following the trauma. Stem cells, due to their capacity to differentiate into neuronal cells and through releasing neurotrophic factors, seem to be a valid strategy to use in the treatment of traumatic brain injury. The purpose of this review is to provide an overview of clinical trials, aimed to evaluate the use of stem cell-based therapy in traumatic brain injury. These studies aim to assess the safety and efficacy of stem cells in this disease. The results available so far are few; therefore, future studies need in order to evaluate the safety and efficacy of stem cell transplantation in traumatic brain injury.
Collapse
|
11
|
Das M, Mayilsamy K, Tang X, Han JY, Foran E, Willing AE, Mohapatra SS, Mohapatra S. Pioglitazone treatment prior to transplantation improves the efficacy of human mesenchymal stem cells after traumatic brain injury in rats. Sci Rep 2019; 9:13646. [PMID: 31541141 PMCID: PMC6754424 DOI: 10.1038/s41598-019-49428-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 08/19/2019] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury is a leading cause of death and disability around the world. So far, drugs are not available to repair brain damage. Human mesenchymal stem cell (hMSC) transplantation therapy is a promising approach, although the inflammatory microenvironment of the injured brain affects the efficacy of transplanted hMSCs. We hypothesize that reducing the inflammation in the cerebral microenvironment by reducing pro-inflammatory chemokines prior to hMSC administration will improve the efficacy of hMSC therapy. In a rat model of lateral fluid percussion injury, combined pioglitazone (PG) and hMSC (combination) treatment showed less anxiety-like behavior and improved sensorimotor responses to a noxious cold stimulus. Significant reduction in brain lesion volume, neurodegeneration, microgliosis and astrogliosis were observed after combination treatment. TBI induced expression of inflammatory chemokine CCL20 and IL1-β were significantly decreased in the combination treatment group. Combination treatment significantly increased brain-derived neurotrophic factor (BDNF) level and subventricular zone (SVZ) neurogenesis. Taken together, reducing proinflammatory cytokine expression in the cerebral tissues after TBI by PG administration and prior to hMSC therapy improves the outcome of the therapy in which BDNF could have a role.
Collapse
Affiliation(s)
- Mahasweta Das
- James A. Haley Veterans Hospital, University of South Florida College of Medicine, Tampa, FL, 33612, USA
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Karthick Mayilsamy
- James A. Haley Veterans Hospital, University of South Florida College of Medicine, Tampa, FL, 33612, USA
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Xiaolan Tang
- James A. Haley Veterans Hospital, University of South Florida College of Medicine, Tampa, FL, 33612, USA
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Jung Yeon Han
- James A. Haley Veterans Hospital, University of South Florida College of Medicine, Tampa, FL, 33612, USA
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Elspeth Foran
- James A. Haley Veterans Hospital, University of South Florida College of Medicine, Tampa, FL, 33612, USA
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Alison E Willing
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Shyam S Mohapatra
- James A. Haley Veterans Hospital, University of South Florida College of Medicine, Tampa, FL, 33612, USA
- Department of Internal Medicine, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Subhra Mohapatra
- James A. Haley Veterans Hospital, University of South Florida College of Medicine, Tampa, FL, 33612, USA.
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL, 33612, USA.
| |
Collapse
|
12
|
Masterson CH, Curley GF, Laffey JG. Modulating the distribution and fate of exogenously delivered MSCs to enhance therapeutic potential: knowns and unknowns. Intensive Care Med Exp 2019; 7:41. [PMID: 31346794 PMCID: PMC6658643 DOI: 10.1186/s40635-019-0235-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 03/07/2019] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are undergoing intensive translational research for several debilitating conditions, including critical illnesses such as ARDS and sepsis. MSCs exert diverse biologic effects via their interaction with host tissues, via mechanisms that require the MSC to be in close proximity to the area of injury. Fully harnessing the therapeutic potential of advanced medicinal therapeutic products such as MSCs and their successful translation to clinical use requires a detailed understanding of MSC distribution and persistence in the injured tissues. Key aspects include understanding MSC distribution within the body, the response of the host to MSC administration, and the ultimate fate of exogenously administered MSCs within the host. Factors affecting this interaction include the MSC tissue source, the in vitro MSC culture conditions, the route of MSC administration and the specific issues relating to the target disease state, each of which remains to be fully characterised. Understanding these factors may generate strategies to modify MSC distribution and fate that may enhance their therapeutic effect. This review will examine our understanding of the mechanisms of action of MSCs, the early and late phase distribution kinetics of MSCs following in vivo administration, the ultimate fate of MSCs following administration and the potential importance of these MSC properties to their therapeutic effects. We will critique current cellular imaging and tracking methodologies used to track exogenous MSCs and their suitability for use in patients, discuss the insights they provide into the distribution and fate of MSCs after administration, and suggest strategies by which MSC biodistribution and fate may be modulated for therapeutic effect and clinical use. In conclusion, a better understanding of patterns of biodistribution and of the fate of MSCs will add important additional safety data regarding MSCs, address regulatory requirements, and may uncover strategies to increase the distribution and/or persistence of MSC at the sites of injury, potentially increasing their therapeutic potential for multiple disorders.
Collapse
Affiliation(s)
- Claire H Masterson
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland.,School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Gerard F Curley
- Department of Anaesthesia and Critical Care, Royal College of Surgeons in Ireland Education and Research Centre Smurfit Building, Beaumont Hospital, Dublin, 9, Ireland
| | - John G Laffey
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland. .,School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland. .,Department of Anaesthesia and Intensive Care Medicine, Galway University Hospitals, SAOLTA Hospital Group, Galway, Ireland.
| |
Collapse
|
13
|
Zhang Y, Chopp M, Rex CS, Simmon VF, Sarraf ST, Zhang ZG, Mahmood A, Xiong Y. A Small Molecule Spinogenic Compound Enhances Functional Outcome and Dendritic Spine Plasticity in a Rat Model of Traumatic Brain Injury. J Neurotrauma 2018; 36:589-600. [PMID: 30014757 DOI: 10.1089/neu.2018.5790] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The tetra (ethylene glycol) derivative of benzothiazole aniline (SPG101) has been shown to improve dendritic spine density and cognitive memory in the triple transgenic mouse model of Alzheimer disease (AD) when administered intraperitoneally. The present study was designed to investigate the therapeutic effects of SPG101 on dendritic spine density and morphology and sensorimotor and cognitive functional recovery in a rat model of traumatic brain injury (TBI) induced by controlled cortical impact (CCI). Young adult male Wistar rats with CCI were randomly divided into the following two groups (n = 7/group): (1) Vehicle, and (2) SPG101. SPG101 (30 mg/kg) dissolved in vehicle (1% dimethyl sulfoxide in phosphate buffered saline) or Vehicle were intraperitoneally administered starting at 1 h post-injury and once daily for the next 34 days. Sensorimotor deficits were assessed using a modified neurological severity score and adhesive removal and foot fault tests. Cognitive function was measured by Morris water maze, novel object recognition (NOR), and three-chamber social recognition tests. The animals were sacrificed 35 days after injury, and their brains were processed for measurement of dendritic spine density and morphology using ballistic dye labeling. Compared with the vehicle treatment, SPG101 treatment initiated 1 h post-injury significantly improved sensorimotor functional recovery (days 7-35, p < 0.0001), spatial learning (days 32-35, p < 0.0001), NOR (days 14 and 35, p < 0.0001), social recognition (days 14 and 35, p < 0.0001). Further, treatment significantly increased dendritic spine density in the injured cortex (p < 0.05), decreased heterogeneous distribution of spine lengths in the injured cortex and hippocampus (p < 0.0001), modifications that are associated with the promotion of spine maturation in these brain regions. In summary, treatment with SPG101 initiated 1 h post-injury and continued for an additional 34 days improves both sensorimotor and cognitive functional recovery, indicating that SPG101 acts as a spinogenic agent and may have potential as a novel treatment of TBI.
Collapse
Affiliation(s)
- Yanlu Zhang
- 1 Department of Neurosurgery, Henry Ford Hospital , Detroit, Michigan
| | - Michael Chopp
- 2 Department of Neurology, Henry Ford Hospital , Detroit, Michigan.,3 Department of Physics, Oakland University , Rochester, Michigan
| | | | | | | | - Zheng Gang Zhang
- 2 Department of Neurology, Henry Ford Hospital , Detroit, Michigan
| | - Asim Mahmood
- 1 Department of Neurosurgery, Henry Ford Hospital , Detroit, Michigan
| | - Ye Xiong
- 1 Department of Neurosurgery, Henry Ford Hospital , Detroit, Michigan
| |
Collapse
|
14
|
Williams AM, Dennahy IS, Bhatti UF, Halaweish I, Xiong Y, Chang P, Nikolian VC, Chtraklin K, Brown J, Zhang Y, Zhang ZG, Chopp M, Buller B, Alam HB. Mesenchymal Stem Cell-Derived Exosomes Provide Neuroprotection and Improve Long-Term Neurologic Outcomes in a Swine Model of Traumatic Brain Injury and Hemorrhagic Shock. J Neurotrauma 2018; 36:54-60. [PMID: 29690826 DOI: 10.1089/neu.2018.5711] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Combined traumatic brain injury (TBI) and hemorrhagic shock (HS) remains a leading cause of preventable death worldwide. Mesenchymal stem cell-derived exosomes have demonstrated promise in small animal models of neurologic injury. To investigate the effects of exosome treatment in a clinically realistic large animal model, Yorkshire swine underwent TBI and HS. Animals were maintained in shock for 2 h before resuscitation with normal saline (NS). Animals were then resuscitated either with NS (3 × volume of shed blood) or with the same volume of NS with delayed exosome administration (1 × 1013 particles/4 mL) (n = 5/cohort). Exosomes were administered 9 h post-injury, and on post-injury days (PID) 1, 5, 9, and 13. Neurologic severity scores (NSS) were assessed for 30 days, and neurocognitive functions were objectively measured. Exosome-treated animals had significantly lower NSS (p < 0.05) during the first five days of recovery. Exosome-treated animals also had a significantly shorter time to complete neurologic recovery (NSS = 0) compared with animals given NS alone (days to recovery: NS = 16.8 ± 10.6; NS + exosomes = 5.6 ± 2.8; p = 0.03). Animals treated with exosomes initiated neurocognitive testing earlier (days to initiation: NS = 9.6 ± 0.5 vs. NS + exosomes = 4.2 ± 0.8; p = 0.008); however, no difference was seen in time to mastery of tasks. In conclusion, treatment with exosomes attenuates the severity of neurologic injury and allows for faster neurologic recovery in a clinically realistic large animal model of TBI and HS.
Collapse
Affiliation(s)
- Aaron M Williams
- 1 Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Isabel S Dennahy
- 1 Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Umar F Bhatti
- 1 Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Ihab Halaweish
- 1 Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Ye Xiong
- 2 Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan
| | - Panpan Chang
- 1 Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Vahagn C Nikolian
- 1 Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Kiril Chtraklin
- 1 Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Jordana Brown
- 1 Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Yanlu Zhang
- 2 Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan
| | - Zheng Gang Zhang
- 3 Department of Neurology, Henry Ford Hospital, Detroit, Michigan
| | - Michael Chopp
- 3 Department of Neurology, Henry Ford Hospital, Detroit, Michigan.,4 Department of Physics, Oakland University, Rochester, Michigan
| | - Benjamin Buller
- 3 Department of Neurology, Henry Ford Hospital, Detroit, Michigan
| | - Hasan B Alam
- 1 Department of Surgery, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
15
|
Xiong Y, Mahmood A, Chopp M. Current understanding of neuroinflammation after traumatic brain injury and cell-based therapeutic opportunities. Chin J Traumatol 2018; 21:137-151. [PMID: 29764704 PMCID: PMC6034172 DOI: 10.1016/j.cjtee.2018.02.003] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 02/04/2023] Open
Abstract
Traumatic brain injury (TBI) remains a major cause of death and disability worldwide. Increasing evidence indicates that TBI is an important risk factor for neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, and chronic traumatic encephalopathy. Despite improved supportive and rehabilitative care of TBI patients, unfortunately, all late phase clinical trials in TBI have yet to yield a safe and effective neuroprotective treatment. The disappointing clinical trials may be attributed to variability in treatment approaches and heterogeneity of the population of TBI patients as well as a race against time to prevent or reduce inexorable cell death. TBI is not just an acute event but a chronic disease. Among many mechanisms involved in secondary injury after TBI, emerging preclinical studies indicate that posttraumatic prolonged and progressive neuroinflammation is associated with neurodegeneration which may be treatable long after the initiating brain injury. This review provides an overview of recent understanding of neuroinflammation in TBI and preclinical cell-based therapies that target neuroinflammation and promote functional recovery after TBI.
Collapse
Affiliation(s)
- Ye Xiong
- Department of Neurosurgery Henry Ford Health System, 2799 West Grand Boulevard, Detroit, MI, 48202, USA.
| | - Asim Mahmood
- Department of Neurosurgery Henry Ford Health System, 2799 West Grand Boulevard, Detroit, MI, 48202, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, 2799 West Grand Boulevard, Detroit, MI, 48202, USA; Department of Physics, Oakland University, Rochester, MI, 48309, USA
| |
Collapse
|
16
|
Park BN, Yoon JK, An YS. Bone marrow mesenchymal stem cell transplantation in acute brain trauma. Nuklearmedizin 2018; 52:192-7. [DOI: 10.3413/nukmed-0543-12-11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 05/06/2013] [Indexed: 01/22/2023]
Abstract
SummaryAim: This study was performed to evaluate the effects of intravenously transplanted rat bone-marrow derived mesenchymal stem cells (rBMSCs) in an acute brain trauma model using serial 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET) in rat models. Animals, methods: Trauma models were made using a controlled cortical impact injury device. The stem cell treatment group was treated with intravenous injections of BMSCs, and models without stem cell therapy comprised the control group. Serial 18F-FDG PET images were obtained 1, 7, 14, 21, and 28 days after trauma. The difference in 18F-FDG uptake between day 1 and each time point after trauma was analyzed with SPM2 (uncorrected p < 0.005). Results: The stem cell treatment group demonstrated significantly higher 18F-FDG uptake in the right parietal region at 14 days after trauma than at 1 day after trauma. An increase in glucose metabolism in the right parietal cortex appeared on days 21 and 28 after trauma in the group without stem cell treatment. The 18F-FDG uptake in the brain was improved over a broader area, including the right parietal and right primary somatosensory cortex, on days 21 and 28 after trauma in the stem cell treatment group compared with the group without stem cell treatment. Conclusion: BMSC therapy in trauma models led to improved glucose metabolism. This result might support the therapeutic effect of stem cells in brain trauma.
Collapse
|
17
|
Zhao Y, Gibb SL, Zhao J, Moore AN, Hylin MJ, Menge T, Xue H, Baimukanova G, Potter D, Johnson EM, Holcomb JB, Cox CS, Dash PK, Pati S. Wnt3a, a Protein Secreted by Mesenchymal Stem Cells Is Neuroprotective and Promotes Neurocognitive Recovery Following Traumatic Brain Injury. Stem Cells 2017; 34:1263-72. [PMID: 26840479 DOI: 10.1002/stem.2310] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 11/25/2015] [Accepted: 12/15/2015] [Indexed: 12/23/2022]
Abstract
Intravenous administration of bone marrow derived mesenchymal stem cells (MSCs) has been shown to reduce blood brain barrier compromise and improve neurocognition following traumatic brain injury (TBI). These effects occur in the absence of engraftment and differentiation of these cells in the injured brain. Recent studies have shown that soluble factors produced by MSCs mediate a number of the therapeutic effects. In this study, we sought to determine if intravenous administration of MSCs (IV-MSCs) could enhance hippocampal neurogenesis following TBI. Our results demonstrate that IV-MSC treatment attenuates loss of neural stem cells and promotes hippocampal neurogenesis in TBI injured mice. As Wnt signaling has been implicated in neurogenesis, we measured circulating Wnt3a levels in serum following IV-MSC administration and found a significant increase in Wnt3a. Concurrent with this increase, we detected increased activation of the Wnt/β-catenin signaling pathway in hippocampal neurons. Furthermore, IV recombinant Wnt3a treatment provided neuroprotection, promoted neurogenesis, and improved neurocognitive function in TBI injured mice. Taken together, our results demonstrate a role for Wnt3a in the therapeutic potential of MSCs and identify Wnt3a as a potential stand-alone therapy or as part of a combination therapeutic strategy for the treatment of TBI. Stem Cells 2016;34:1263-1272.
Collapse
Affiliation(s)
- Yuhai Zhao
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Stuart L Gibb
- Blood Systems Research Institute, San Francisco, California, USA.,Department of Laboratory Medicine, University of California San Francisco, California, USA
| | - Jing Zhao
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Anthony N Moore
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Michael J Hylin
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, Texas, USA.,Department of Psychology, Southern Illinois University, Carbondale, Illinois, USA
| | - Tyler Menge
- Blood Systems Research Institute, San Francisco, California, USA
| | - Hasen Xue
- Department of Surgery and Center for Translational Injury Research, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Gyulnar Baimukanova
- Blood Systems Research Institute, San Francisco, California, USA.,Department of Laboratory Medicine, University of California San Francisco, California, USA
| | - Daniel Potter
- Blood Systems Research Institute, San Francisco, California, USA.,Department of Laboratory Medicine, University of California San Francisco, California, USA
| | - Evan M Johnson
- Department of Surgery and Center for Translational Injury Research, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - John B Holcomb
- Department of Surgery and Center for Translational Injury Research, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Charles S Cox
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Pramod K Dash
- Blood Systems Research Institute, San Francisco, California, USA
| | - Shibani Pati
- Department of Surgery and Center for Translational Injury Research, University of Texas Health Science Center at Houston, Houston, Texas, USA.,Department of Psychology, Southern Illinois University, Carbondale, Illinois, USA
| |
Collapse
|
18
|
Li Y, Yang YY, Ren JL, Xu F, Chen FM, Li A. Exosomes secreted by stem cells from human exfoliated deciduous teeth contribute to functional recovery after traumatic brain injury by shifting microglia M1/M2 polarization in rats. Stem Cell Res Ther 2017; 8:198. [PMID: 28962585 PMCID: PMC5622448 DOI: 10.1186/s13287-017-0648-5] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 08/11/2017] [Accepted: 08/14/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is one of the major causes of mortality and disability for all ages worldwide. Mesenchymal stem cells (MSCs)-originated exosomes have provided therapeutic effects. However, as an indispensable component of MSCs, whether odontogenic stem cell-generated exosomes could benefit TBI is still unclear. Thus we aimed to explore the potential of stem cells from human exfoliated deciduous teeth-originated exosomes (SHED-Ex) for the management of TBI. METHODS First, a transwell system was used to co-culture activated BV-2 microglia cells with SHED. The secretion levels of neuroinflammatory factors and nitrite were evaluated by enzyme-linked immunosorbent assay (ELISA) and Griess assay. Furthermore, purified SHED-Ex were co-cultured with activated BV-2. ELISA, Griess assay, flow cytometry, immunofluorescence, and qRT-PCR were performed to test the levels of inflammatory factors as well as the microglia phenotype. Finally, SHED and SHED-Ex were locally injected into TBI rat models. Basso, Beattie, and Bresnahan (BBB) scores were chosen to evaluate the motor functional recovery. Histopathology and immunofluorescence were performed to measure the lesion volume and neuroinflammation. RESULTS As a result, SHED-Ex could reduce neuroinflammation by shifting microglia polarization. The administration of SHED-Ex improves rat motor functional recovery and reduces cortical lesion compared with the control group 2 weeks post-injury (P < 0.05). CONCLUSIONS The current study demonstrates for the first time that SHED-Ex contribute a therapeutic benefit to TBI in rats, at least in part by shifting microglia polarization to reduce neuroinflammation. The use of odontogenic stem cells, and indeed their exosomes, may be expanded for the treatment of TBI or other neurological disorders.
Collapse
Affiliation(s)
- Ye Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Xi'an, Shaanxi, China.,Department of Periodontology, College of Stomatology, Xi'an Jiaotong University, Xi Wu Road No.98, Xi'an, Shaanxi, 710004, China
| | - Yuan-Yuan Yang
- Department of Periodontology, College of Stomatology, Xi'an Jiaotong University, Xi Wu Road No.98, Xi'an, Shaanxi, 710004, China
| | - Jia-Li Ren
- Department of Periodontology, College of Stomatology, Xi'an Jiaotong University, Xi Wu Road No.98, Xi'an, Shaanxi, 710004, China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University School of Life Science and Technology, Xi'an, China.,Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, China
| | - Fa-Ming Chen
- Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Ang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Xi'an, Shaanxi, China. .,Department of Periodontology, College of Stomatology, Xi'an Jiaotong University, Xi Wu Road No.98, Xi'an, Shaanxi, 710004, China.
| |
Collapse
|
19
|
Boda E, Nato G, Buffo A. Emerging pharmacological approaches to promote neurogenesis from endogenous glial cells. Biochem Pharmacol 2017. [PMID: 28647491 DOI: 10.1016/j.bcp.2017.06.129] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Neurodegenerative disorders are emerging as leading contributors to the global disease burden. While some drug-based approaches have been designed to limit or prevent neuronal loss following acute damage or chronic neurodegeneration, regeneration of functional neurons in the adult Central Nervous System (CNS) still remains an unmet need. In this context, the exploitation of endogenous cell sources has recently gained an unprecedented attention, thanks to the demonstration that, in some CNS regions or under specific circumstances, glial cells can activate spontaneous neurogenesis or can be instructed to produce neurons in the adult mammalian CNS parenchyma. This field of research has greatly advanced in the last years and identified interesting molecular and cellular mechanisms guiding the neurogenic activation/conversion of glia. In this review, we summarize the evolution of the research devoted to understand how resident glia can be directed to produce neurons. We paid particular attention to pharmacologically-relevant approaches exploiting the modulation of niche-associated factors and the application of selected small molecules.
Collapse
Affiliation(s)
- Enrica Boda
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, I-10126 Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi, I-10043 Orbassano, Turin, Italy.
| | - Giulia Nato
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, I-10126 Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi, I-10043 Orbassano, Turin, Italy
| | - Annalisa Buffo
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, I-10126 Turin, Italy; Neuroscience Institute Cavalieri Ottolenghi, I-10043 Orbassano, Turin, Italy
| |
Collapse
|
20
|
Geddes RI, Hayashi K, Bongers Q, Wehber M, Anderson IM, Jansen AD, Nier C, Fares E, Farquhar G, Kapoor A, Ziegler TE, VadakkadathMeethal S, Bird IM, Atwood CS. Conjugated Linoleic Acid Administration Induces Amnesia in Male Sprague Dawley Rats and Exacerbates Recovery from Functional Deficits Induced by a Controlled Cortical Impact Injury. PLoS One 2017; 12:e0169494. [PMID: 28125600 PMCID: PMC5268708 DOI: 10.1371/journal.pone.0169494] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 12/16/2016] [Indexed: 12/05/2022] Open
Abstract
Long-chain polyunsaturated fatty acids like conjugated linoleic acids (CLA) are required for normal neural development and cognitive function and have been ascribed various beneficial functions. Recently, oral CLA also has been shown to increase testosterone (T) biosynthesis, which is known to diminish traumatic brain injury (TBI)-induced neuropathology and reduce deficits induced by stroke in adult rats. To test the impact of CLA on cognitive recovery following a TBI, 5-6 month old male Sprague Dawley rats received a focal injury (craniectomy + controlled cortical impact (CCI; n = 17)) or Sham injury (craniectomy alone; n = 12) and were injected with 25 mg/kg body weight of Clarinol® G-80 (80% CLA in safflower oil; n = 16) or saline (n = 13) every 48 h for 4 weeks. Sham surgery decreased baseline plasma progesterone (P4) by 64.2% (from 9.5 ± 3.4 ng/mL to 3.4 ± 0.5 ng/mL; p = 0.068), T by 74.6% (from 5.9 ± 1.2 ng/mL to 1.5 ± 0.3 ng/mL; p < 0.05), 11-deoxycorticosterone (11-DOC) by 37.5% (from 289.3 ± 42.0 ng/mL to 180.7 ± 3.3 ng/mL), and corticosterone by 50.8% (from 195.1 ± 22.4 ng/mL to 95.9 ± 2.2 ng/mL), by post-surgery day 1. CCI injury induced similar declines in P4, T, 11-DOC and corticosterone (58.9%, 74.6%, 39.4% and 24.6%, respectively) by post-surgery day 1. These results suggest that both Sham surgery and CCI injury induce hypogonadism and hypoadrenalism in adult male rats. CLA treatment did not reverse hypogonadism in Sham (P4: 2.5 ± 1.0 ng/mL; T: 0.9 ± 0.2 ng/mL) or CCI-injured (P4: 2.2 ± 0.9 ng/mL; T: 1.0 ± 0.2 ng/mL, p > 0.05) animals by post-injury day 29, but rapidly reversed by post-injury day 1 the hypoadrenalism in Sham (11-DOC: 372.6 ± 36.6 ng/mL; corticosterone: 202.6 ± 15.6 ng/mL) and CCI-injured (11-DOC: 384.2 ± 101.3 ng/mL; corticosterone: 234.6 ± 43.8 ng/mL) animals. In Sham surgery animals, CLA did not alter body weight, but did markedly increase latency to find the hidden Morris Water Maze platform (40.3 ± 13.0 s) compared to saline treated Sham animals (8.8 ± 1.7 s). In CCI injured animals, CLA did not alter CCI-induced body weight loss, CCI-induced cystic infarct size, or deficits in rotarod performance. However, like Sham animals, CLA injections exacerbated the latency of CCI-injured rats to find the hidden MWM platform (66.8 ± 10.6 s) compared to CCI-injured rats treated with saline (30.7 ± 5.5 s, p < 0.05). These results indicate that chronic treatment of CLA at a dose of 25 mg/kg body weight in adult male rats over 1-month 1) does not reverse craniectomy- and craniectomy + CCI-induced hypogonadism, but does reverse craniectomy- and craniectomy + CCI-induced hypoadrenalism, 2) is detrimental to medium- and long-term spatial learning and memory in craniectomized uninjured rats, 3) limits cognitive recovery following a moderate-severe CCI injury, and 4) does not alter body weight.
Collapse
Affiliation(s)
- Rastafa I. Geddes
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Kentaro Hayashi
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Quinn Bongers
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Marlyse Wehber
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Icelle M. Anderson
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Alex D. Jansen
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Chase Nier
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Emily Fares
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Gabrielle Farquhar
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Amita Kapoor
- Assay Services Unit and Institute for Clinical and Translational Research Core Laboratory, National Primate Research Center, University of Wisconsin-Madison, Wisconsin, United States of America
| | - Toni E. Ziegler
- Assay Services Unit and Institute for Clinical and Translational Research Core Laboratory, National Primate Research Center, University of Wisconsin-Madison, Wisconsin, United States of America
| | - Sivan VadakkadathMeethal
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Ian M. Bird
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Craig S. Atwood
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
- Geriatric Research, Education and Clinical Center, Veterans Administration Hospital, Madison, Wisconsin, United States of America
- School of Exercise, Biomedical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| |
Collapse
|
21
|
Guo S, Zhen Y, Wang A. Transplantation of bone mesenchymal stem cells promotes angiogenesis and improves neurological function after traumatic brain injury in mouse. Neuropsychiatr Dis Treat 2017; 13:2757-2765. [PMID: 29158675 PMCID: PMC5683767 DOI: 10.2147/ndt.s141534] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Traumatic brain injury (TBI) has emerged as a leading cause of mortality and morbidity worldwide. Transplantation of bone mesenchymal stem cells (BMSCs) has emerged as a promising treatment for various central nervous system diseases. This study aims to evaluate the effect of BMSCs transplantation by intravenous injection on neurological function and angiogenesis of the TBI mice. C57BL/6 male mice were randomly divided into four groups: control, sham, TBI, and BMSC. Functional neurological evaluation was performed 1, 4, 7, 14, and 21 days after TBI using neurological severity scores. The impairment of learning and memory in mice was evaluated 14 days after TBI by Morris water maze experiment. Mice were sacrificed 14 days after TBI, and then brain sections were stained by terminal deoxyribonucleotidyl transferase (TDT)-mediated dUTP-digoxigenin nick end labeling staining to assess brain neuronal apoptosis. Immunohistochemistry was conducted to evaluate caspase-3 activity and identify vascular distribution and microvessel density. Protein and mRNA levels of vascular endothelial growth factor (VEGF) and angiogenin-1 (Ang-1) in brain tissues were analyzed by Western blot and quantitative real-time polymerase chain reaction, respectively. BMSCs transplantation promoted recovery of neurological function, ameliorated impairment of learning and memory, attenuated neuronal apoptosis, and diminished caspase-3 activation in mice after TBI. Also, BMSCs transplantation upregulated expressions of VEGF and Ang-1 and promoted the formation of microvessels in brain tissues after TBI. Our study demonstrated the important role of BMSCs transplantation in TBI mouse and indicated that the underlying mechanism was through promoting angiogenesis and improving neurological function. This provides a novel and effective strategy for cell-based therapy in the treatment of TBI.
Collapse
Affiliation(s)
- Shewei Guo
- Department of Neurosurgery, The First Affiliated Hospital, Zhengzhou University, Henan, China
| | - Yingwei Zhen
- Department of Neurosurgery, The First Affiliated Hospital, Zhengzhou University, Henan, China
| | - Anran Wang
- Department of Neurosurgery, The First Affiliated Hospital, Zhengzhou University, Henan, China
| |
Collapse
|
22
|
Moore TL, Pessina MA, Finklestein SP, Killiany RJ, Bowley B, Benowitz L, Rosene DL. Inosine enhances recovery of grasp following cortical injury to the primary motor cortex of the rhesus monkey. Restor Neurol Neurosci 2016; 34:827-48. [PMID: 27497459 PMCID: PMC6503840 DOI: 10.3233/rnn-160661] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Inosine, a naturally occurring purine nucleoside, has been shown to stimulate axonal growth in cell culture and promote corticospinal tract axons to sprout collateral branches after stroke, spinal cord injury and TBI in rodent models. OBJECTIVE To explore the effects of inosine on the recovery of motor function following cortical injury in the rhesus monkey. METHODS After being trained on a test of fine motor function of the hand, monkeys received a lesion limited to the area of the hand representation in primary motor cortex. Beginning 24 hours after this injury and continuing daily thereafter, monkeys received orally administered inosine (500 mg) or placebo. Retesting of motor function began on the 14th day after injury and continued for 12 weeks. RESULTS During the first 14 days after surgery, there was evidence of significant recovery within the inosine-treated group on measures of fine motor function of the hand, measures of hand strength and digit flexion. While there was no effect of treatment on the time to retrieve a reward, the treated monkeys returned to asymptotic levels of grasp performance significantly faster than the untreated monkeys. Additionally, the treated monkeys evidenced a greater degree of recovery in terms of maturity of grasp pattern. CONCLUSION These findings demonstrate that inosine can enhance recovery of function following cortical injury in monkeys.
Collapse
Affiliation(s)
- Tara L. Moore
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Monica A. Pessina
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | | | - Ronald J. Killiany
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Bethany Bowley
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Larry Benowitz
- Department of Neurosurgery and F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Douglas L. Rosene
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| |
Collapse
|
23
|
Zhang Y, Chopp M, Zhang ZG, Katakowski M, Xin H, Qu C, Ali M, Mahmood A, Xiong Y. Systemic administration of cell-free exosomes generated by human bone marrow derived mesenchymal stem cells cultured under 2D and 3D conditions improves functional recovery in rats after traumatic brain injury. Neurochem Int 2016; 111:69-81. [PMID: 27539657 DOI: 10.1016/j.neuint.2016.08.003] [Citation(s) in RCA: 284] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/22/2016] [Accepted: 08/10/2016] [Indexed: 12/13/2022]
Abstract
Multipotent human bone marrow derived mesenchymal stem cells (hMSCs) improve functional outcome after experimental traumatic brain injury (TBI). The present study was designed to investigate whether systemic administration of cell-free exosomes generated from hMSCs cultured in 2-dimensional (2D) conventional conditions or in 3-dimensional (3D) collagen scaffolds promote functional recovery and neurovascular remodeling in rats after TBI. Wistar rats were subjected to TBI induced by controlled cortical impact; 24 h later tail vein injection of exosomes derived from hMSCs cultured under 2D or 3D conditions or an equal number of liposomes as a treatment control were performed. The modified Morris water maze, neurological severity score and footfault tests were employed to evaluate cognitive and sensorimotor functional recovery. Animals were sacrificed at 35 days after TBI. Histological and immunohistochemical analyses were performed for measurements of lesion volume, neurovascular remodeling (angiogenesis and neurogenesis), and neuroinflammation. Compared with liposome-treated control, exosome-treatments did not reduce lesion size but significantly improved spatial learning at 33-35 days measured by the Morris water maze test, and sensorimotor functional recovery, i.e., reduced neurological deficits and footfault frequency, observed at 14-35 days post injury (p < 0.05). Exosome treatments significantly increased the number of newborn endothelial cells in the lesion boundary zone and dentate gyrus, and significantly increased the number of newborn mature neurons in the dentate gyrus as well as reduced neuroinflammation. Exosomes derived from hMSCs cultured in 3D scaffolds provided better outcome in spatial learning than exosomes from hMSCs cultured in the 2D condition. In conclusion, hMSC-generated exosomes significantly improve functional recovery in rats after TBI, at least in part, by promoting endogenous angiogenesis and neurogenesis and reducing neuroinflammation. Thus, exosomes derived from hMSCs may be a novel cell-free therapy for TBI, and hMSC-scaffold generated exosomes may selectively enhance spatial learning.
Collapse
Affiliation(s)
- Yanlu Zhang
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA; Department of Physics, Oakland University, Rochester, MI, USA
| | | | - Mark Katakowski
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Hongqi Xin
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Changsheng Qu
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, USA
| | - Meser Ali
- Department of Radiology, Henry Ford Hospital, Detroit, MI, USA
| | - Asim Mahmood
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, USA
| | - Ye Xiong
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, USA.
| |
Collapse
|
24
|
Gibb SL, Zhao Y, Potter D, Hylin MJ, Bruhn R, Baimukanova G, Zhao J, Xue H, Abdel-Mohsen M, Pillai SK, Moore AN, Johnson EM, Cox CS, Dash PK, Pati S. TIMP3 Attenuates the Loss of Neural Stem Cells, Mature Neurons and Neurocognitive Dysfunction in Traumatic Brain Injury. Stem Cells 2015; 33:3530-44. [PMID: 26299440 DOI: 10.1002/stem.2189] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 07/30/2015] [Accepted: 08/07/2015] [Indexed: 12/19/2022]
Abstract
Mesenchymal stem cells (MSCs) have been shown to have potent therapeutic effects in a number of disorders including traumatic brain injury (TBI). However, the molecular mechanism(s) underlying these protective effects are largely unknown. Herein we demonstrate that tissue inhibitor of matrix metalloproteinase-3 (TIMP3), a soluble protein released by MSCs, is neuroprotective and enhances neuronal survival and neurite outgrowth in vitro. In vivo in a murine model of TBI, intravenous recombinant TIMP3 enhances dendritic outgrowth and abrogates loss of hippocampal neural stem cells and mature neurons. Mechanistically we demonstrate in vitro and in vivo that TIMP3-mediated neuroprotection is critically dependent on activation of the Akt-mTORC1 pathway. In support of the neuroprotective effect of TIMP3, we find that intravenous delivery of recombinant TIMP3 attenuates deficits in hippocampal-dependent neurocognition. Taken together, our data strongly suggest that TIMP3 has direct neuroprotective effects that can mitigate the deleterious effects associated with TBI, an area with few if any therapeutic options.
Collapse
Affiliation(s)
- Stuart L Gibb
- Blood Systems Research Institute, San Francisco, California, USA.,Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Yuhai Zhao
- Department of Neurobiology and Anatomy, The University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Daniel Potter
- Blood Systems Research Institute, San Francisco, California, USA.,Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Michael J Hylin
- Department of Neurobiology and Anatomy, The University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Roberta Bruhn
- Blood Systems Research Institute, San Francisco, California, USA.,Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Gyulnar Baimukanova
- Blood Systems Research Institute, San Francisco, California, USA.,Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jing Zhao
- Department of Neurobiology and Anatomy, The University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Hasen Xue
- Department of Pediatric Surgery and Institute for Molecular Medicine, The University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Mohamed Abdel-Mohsen
- Blood Systems Research Institute, San Francisco, California, USA.,Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Satish K Pillai
- Blood Systems Research Institute, San Francisco, California, USA.,Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Anthony N Moore
- Department of Neurobiology and Anatomy, The University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Evan M Johnson
- Department of Neurobiology and Anatomy, The University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Charles S Cox
- Department of Pediatric Surgery and Institute for Molecular Medicine, The University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Pramod K Dash
- Department of Neurobiology and Anatomy, The University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Shibani Pati
- Blood Systems Research Institute, San Francisco, California, USA.,Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
25
|
Gennai S, Monsel A, Hao Q, Liu J, Gudapati V, Barbier EL, Lee JW. Cell-based therapy for traumatic brain injury. Br J Anaesth 2015; 115:203-12. [PMID: 26170348 DOI: 10.1093/bja/aev229] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Traumatic brain injury is a major economic burden to hospitals in terms of emergency department visits, hospitalizations, and utilization of intensive care units. Current guidelines for the management of severe traumatic brain injuries are primarily supportive, with an emphasis on surveillance (i.e. intracranial pressure) and preventive measures to reduce morbidity and mortality. There are no direct effective therapies available. Over the last fifteen years, pre-clinical studies in regenerative medicine utilizing cell-based therapy have generated enthusiasm as a possible treatment option for traumatic brain injury. In these studies, stem cells and progenitor cells were shown to migrate into the injured brain and proliferate, exerting protective effects through possible cell replacement, gene and protein transfer, and release of anti-inflammatory and growth factors. In this work, we reviewed the pathophysiological mechanisms of traumatic brain injury, the biological rationale for using stem cells and progenitor cells, and the results of clinical trials using cell-based therapy for traumatic brain injury. Although the benefits of cell-based therapy have been clearly demonstrated in pre-clinical studies, some questions remain regarding the biological mechanisms of repair and safety, dose, route and timing of cell delivery, which ultimately will determine its optimal clinical use.
Collapse
Affiliation(s)
- S Gennai
- Department of Emergency Medicine, Grenoble University Hospital, La Tronche, France
| | - A Monsel
- Multidisciplinary Intensive Care Unit, Department of Anesthesiology and Critical Care, La Pitié-Salpêtrière Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Q Hao
- Department of Anesthesiology, University of California San Francisco, 505 Parnassus Ave., Box 0648, San Francisco, CA 94143, USA
| | - J Liu
- Department of Anesthesiology, University of California San Francisco, 505 Parnassus Ave., Box 0648, San Francisco, CA 94143, USA
| | - V Gudapati
- Department of Anesthesiology, University of California San Francisco, 505 Parnassus Ave., Box 0648, San Francisco, CA 94143, USA
| | - E L Barbier
- Grenoble Institut des Neurosciences, Unité Inserm U 836, La Tronche, France
| | - J W Lee
- Department of Anesthesiology, University of California San Francisco, 505 Parnassus Ave., Box 0648, San Francisco, CA 94143, USA
| |
Collapse
|
26
|
Gabriel N, Samuel R, Jayandharan GR. Targeted delivery of AAV-transduced mesenchymal stromal cells to hepatic tissue forex vivogene therapy. J Tissue Eng Regen Med 2015; 11:1354-1364. [DOI: 10.1002/term.2034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/02/2015] [Accepted: 04/21/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Nishanth Gabriel
- Department of Haematology; Christian Medical College; Vellore Tamil Nadu India
| | - Rekha Samuel
- Centre for Stem Cell Research; Christian Medical College; Vellore Tamil Nadu India
| | - Giridhara R. Jayandharan
- Department of Haematology; Christian Medical College; Vellore Tamil Nadu India
- Department of Biological Sciences and Bioengineering; Indian Institute of Technology; Kanpur Uttar Pradesh India
| |
Collapse
|
27
|
Xiong Y, Zhang Y, Mahmood A, Chopp M. Investigational agents for treatment of traumatic brain injury. Expert Opin Investig Drugs 2015; 24:743-60. [PMID: 25727893 PMCID: PMC4433440 DOI: 10.1517/13543784.2015.1021919] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Traumatic brain injury (TBI) is a major cause of death and disability worldwide. To date, there are no pharmacologic agents proven to improve outcomes from TBI because all the Phase III clinical trials in TBI have failed. Thus, there is a compelling need to develop treatments for TBI. AREAS COVERED The following article provides an overview of select cell-based and pharmacological therapies under early development for the treatment of TBI. These therapies seek to enhance cognitive and neurological functional recovery through neuroprotective and neurorestorative strategies. EXPERT OPINION TBI elicits both complex degenerative and regenerative tissue responses in the brain. TBI can lead to cognitive, behavioral, and motor deficits. Although numerous promising neuroprotective treatment options have emerged from preclinical studies that mainly target the lesion, translation of preclinical effective neuroprotective drugs to clinical trials has proven challenging. Accumulating evidence indicates that the mammalian brain has a significant, albeit limited, capacity for both structural and functional plasticity, as well as regeneration essential for spontaneous functional recovery after injury. A new therapeutic approach is to stimulate neurovascular remodeling by enhancing angiogenesis, neurogenesis, oligodendrogenesis, and axonal sprouting, which in concert, may improve neurological functional recovery after TBI.
Collapse
Affiliation(s)
- Ye Xiong
- Henry Ford Hospital, Department of Neurosurgery , Education and Research Building, Room 3096, 2799 West Grand Boulevard, Detroit, MI 48202 , USA +1 313 916 4743 ; +1 313 916 9855 ;
| | | | | | | |
Collapse
|
28
|
Turtzo LC, Budde MD, Dean DD, Gold EM, Lewis BK, Janes L, Lescher J, Coppola T, Yarnell A, Grunberg NE, Frank JA. Failure of intravenous or intracardiac delivery of mesenchymal stromal cells to improve outcomes after focal traumatic brain injury in the female rat. PLoS One 2015; 10:e0126551. [PMID: 25946089 PMCID: PMC4422703 DOI: 10.1371/journal.pone.0126551] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/03/2015] [Indexed: 01/16/2023] Open
Abstract
Mesenchymal stromal cells secrete a variety of anti-inflammatory factors and may provide a regenerative medicine option for the treatment of traumatic brain injury. The present study investigates the efficacy of multiple intravenous or intracardiac administrations of rat mesenchymal stromal cells or human mesenchymal stromal cells in female rats after controlled cortical impact by in vivo MRI, neurobehavior, and histopathology evaluation. Neither intravenous nor intracardiac administration of mesenchymal stromal cells derived from either rats or humans improved MRI measures of lesion volume or neurobehavioral outcome compared to saline treatment. Few mesenchymal stromal cells (<0.0005% of injected dose) were found within 3 days of last dosage at the site of injury after either delivery route, with no mesenchymal stromal cells being detectable in brain at 30 or 56 days post-injury. These findings suggest that non-autologous mesenchymal stromal cells therapy via intravenous or intracardiac administration is not a promising treatment after focal contusion traumatic brain injury in this female rodent model.
Collapse
Affiliation(s)
- L. Christine Turtzo
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- Frank Laboratory, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| | - Matthew D. Budde
- Frank Laboratory, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Dana D. Dean
- Frank Laboratory, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Eric M. Gold
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- Frank Laboratory, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Bobbi K. Lewis
- Frank Laboratory, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lindsay Janes
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- Frank Laboratory, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jacob Lescher
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- Frank Laboratory, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Tiziana Coppola
- Frank Laboratory, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Angela Yarnell
- Department of Medical and Clinical Psychology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Neil E. Grunberg
- Department of Medical and Clinical Psychology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Joseph A. Frank
- Frank Laboratory, National Institutes of Health, Bethesda, Maryland, United States of America
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
29
|
Zhao J, Chen N, Shen N, Zhao H, Wang D, Shi J, Wang Y, Cui X, Yan Z, Xue H. Transplantation of human umbilical cord blood mesenchymal stem cells to treat a rat model of traumatic brain injury. Neural Regen Res 2015; 7:741-8. [PMID: 25737696 PMCID: PMC4345655 DOI: 10.3969/j.issn.1673-5374.2012.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 02/04/2012] [Indexed: 12/13/2022] Open
Abstract
In the present study, human umbilical cord blood mesenchymal stem cells were injected into a rat model of traumatic brain injury via the tail vein. Results showed that 5-bromodeoxyuridine-labeled cells aggregated around the injury site, surviving up to 4 weeks post-transplantation. In addition, transplantation-related death did not occur, and neurological functions significantly improved. Histological detection revealed attenuated pathological injury in rat brain tissues following human umbilical cord blood mesenchymal stem cell transplantation. In addition, the number of apoptotic cells decreased. Immunohistochemistry and in situ hybridization showed increased expression of brain-derived neurotrophic factor, nerve growth factor, basic fibroblast growth factor, and vascular endothelial growth factor, along with increased microvessel density in surrounding areas of brain injury. Results demonstrated migration of transplanted human umbilical cord blood mesenchymal stem cells into the lesioned boundary zone of rats, as well as increased angiogenesis and expression of related neurotrophic factors in the lesioned boundary zone.
Collapse
Affiliation(s)
- Junjian Zhao
- Affiliated Hospital of Hebei United University, Tangshan 063000, Hebei Province, China
| | - Naiyao Chen
- Affiliated Hospital of Hebei United University, Tangshan 063000, Hebei Province, China
| | - Na Shen
- Affiliated Hospital of Hebei United University, Tangshan 063000, Hebei Province, China
| | - Hui Zhao
- Affiliated Hospital of Hebei United University, Tangshan 063000, Hebei Province, China ; Tianjin Key Laboratory of Food Biotechnology, Tianjin University of Commerce, Tianjin 300134, China
| | - Dali Wang
- Affiliated Hospital of Hebei United University, Tangshan 063000, Hebei Province, China
| | - Jun Shi
- Affiliated Hospital of Hebei United University, Tangshan 063000, Hebei Province, China
| | - Yang Wang
- College of Life Science, Hebei United University, Tangshan 063000, Hebei Province, China
| | - Xiufeng Cui
- Affiliated Hospital of Hebei United University, Tangshan 063000, Hebei Province, China
| | - Zhenyu Yan
- Affiliated Hospital of Hebei United University, Tangshan 063000, Hebei Province, China
| | - Hui Xue
- Affiliated Hospital of Hebei United University, Tangshan 063000, Hebei Province, China
| |
Collapse
|
30
|
Cell-based therapy for acute organ injury: preclinical evidence and ongoing clinical trials using mesenchymal stem cells. Anesthesiology 2014; 121:1099-121. [PMID: 25211170 DOI: 10.1097/aln.0000000000000446] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Critically ill patients often suffer from multiple organ failures involving lung, kidney, liver, or brain. Genomic, proteomic, and metabolomic approaches highlight common injury mechanisms leading to acute organ failure. This underlines the need to focus on therapeutic strategies affecting multiple injury pathways. The use of adult stem cells such as mesenchymal stem or stromal cells (MSC) may represent a promising new therapeutic approach as increasing evidence shows that MSC can exert protective effects following injury through the release of promitotic, antiapoptotic, antiinflammatory, and immunomodulatory soluble factors. Furthermore, they can mitigate metabolomic and oxidative stress imbalance. In this work, the authors review the biological capabilities of MSC and the results of clinical trials using MSC as therapy in acute organ injuries. Although preliminary results are encouraging, more studies concerning safety and efficacy of MSC therapy are needed to determine their optimal clinical use. (ANESTHESIOLOGY 2014; 121:1099-121).
Collapse
|
31
|
Skardelly M, Gaber K, Burdack S, Scheidt F, Schuhmann MU, Hilbig H, Meixensberger J, Boltze J. Transient but not permanent benefit of neuronal progenitor cell therapy after traumatic brain injury: potential causes and translational consequences. Front Cell Neurosci 2014; 8:318. [PMID: 25352780 PMCID: PMC4196631 DOI: 10.3389/fncel.2014.00318] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 09/23/2014] [Indexed: 01/16/2023] Open
Abstract
Background: Numerous studies have reported a beneficial impact of neural progenitor cell transplantation on functional outcome after traumatic brain injury (TBI) during short and medium follow-up periods. However, our knowledge regarding long-term functional effects is fragmentary while a direct comparison between local and systemic transplantation is missing so far. Objectives: This study investigated the long-term (12 week) impact of human fetal neuronal progenitor cell (hNPC) transplantation 24 h after severe TBI in rats. Methods: Cells were either transplanted stereotactically (1 × 105) into the putamen or systemically (5 × 105) via the tail vein. Control animals received intravenous transplantation of vehicle solution. Results: An overall functional benefit was observed after systemic, but not local hNPC transplantation by area under the curve analysis (p < 0.01). Surprisingly, this effect vanished during later stages after TBI with all groups exhibiting comparable functional outcomes 84 days after TBI. Investigation of cell-mediated inflammatory processes revealed increasing microglial activation and macrophage presence during these stages, which was statistically significant after systemic cell administration (p < 0.05). Intracerebral hNPC transplantation slightly diminished astrogliosis in perilesional areas (p < 0.01), but did not translate into a permanent functional benefit. No significant effects on angiogenesis were observed among the groups. Conclusion: Our results suggest the careful long-term assessment of cell therapies for TBI, as well as to identify potential long-term detrimental effects of such therapies before moving on to clinical trials. Moreover, immunosuppressive protocols, though widely used, should be rigorously assessed for their applicability in the respective setup.
Collapse
Affiliation(s)
- Marco Skardelly
- Department of Neurosurgery, University of Leipzig Leipzig, Germany
| | - Khaled Gaber
- Department of Neurosurgery, University of Leipzig Leipzig, Germany
| | - Swen Burdack
- Department of Neurosurgery, University of Leipzig Leipzig, Germany
| | | | | | | | | | - Johannes Boltze
- Fraunhofer Institute for Cell Therapy and Immunology and Translational Centre for Regenerative Medicine, University of Leipzig Leipzig, Germany ; Stroke and Neurovascular Regulation Laboratory, Massachussets General Hospital and Harvard Medical School Charlestown, MA, USA
| |
Collapse
|
32
|
Anbari F, Khalili MA, Bahrami AR, Khoradmehr A, Sadeghian F, Fesahat F, Nabi A. Intravenous transplantation of bone marrow mesenchymal stem cells promotes neural regeneration after traumatic brain injury. Neural Regen Res 2014; 9:919-23. [PMID: 25206912 PMCID: PMC4146223 DOI: 10.4103/1673-5374.133133] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2014] [Indexed: 02/06/2023] Open
Abstract
To investigate the supplement of lost nerve cells in rats with traumatic brain injury by intravenous administration of allogenic bone marrow mesenchymal stem cells, this study established a Wistar rat model of traumatic brain injury by weight drop impact acceleration method and administered 3 × 106 rat bone marrow mesenchymal stem cells via the lateral tail vein. At 14 days after cell transplantation, bone marrow mesenchymal stem cells differentiated into neurons and astrocytes in injured rat cerebral cortex and rat neurological function was improved significantly. These findings suggest that intravenously administered bone marrow mesenchymal stem cells can promote nerve cell regeneration in injured cerebral cortex, which supplement the lost nerve cells.
Collapse
Affiliation(s)
- Fatemeh Anbari
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Ali Khalili
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ahmad Reza Bahrami
- Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Arezoo Khoradmehr
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Sadeghian
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Farzaneh Fesahat
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ali Nabi
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
33
|
Skop NB, Calderon F, Cho CH, Gandhi CD, Levison SW. Improvements in biomaterial matrices for neural precursor cell transplantation. MOLECULAR AND CELLULAR THERAPIES 2014; 2:19. [PMID: 26056586 PMCID: PMC4452047 DOI: 10.1186/2052-8426-2-19] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 06/05/2014] [Indexed: 12/24/2022]
Abstract
Progress is being made in developing neuroprotective strategies for traumatic brain injuries; however, there will never be a therapy that will fully preserve neurons that are injured from moderate to severe head injuries. Therefore, to restore neurological function, regenerative strategies will be required. Given the limited regenerative capacity of the resident neural precursors of the CNS, many investigators have evaluated the regenerative potential of transplanted precursors. Unfortunately, these precursors do not thrive when engrafted without a biomaterial scaffold. In this article we review the types of natural and synthetic materials that are being used in brain tissue engineering applications for traumatic brain injury and stroke. We also analyze modifications of the scaffolds including immobilizing drugs, growth factors and extracellular matrix molecules to improve CNS regeneration and functional recovery. We conclude with a discussion of some of the challenges that remain to be solved towards repairing and regenerating the brain.
Collapse
Affiliation(s)
- Nolan B Skop
- Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA ; Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102 USA
| | - Frances Calderon
- Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA
| | - Cheul H Cho
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102 USA
| | - Chirag D Gandhi
- Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA ; Department of Neurological Surgery, Rutgers University-New Jersey Medical School, New Jersey Medical School, Newark, NJ 07103 USA
| | - Steven W Levison
- Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA
| |
Collapse
|
34
|
Skop NB, Calderon F, Cho CH, Gandhi CD, Levison SW. Improvements in biomaterial matrices for neural precursor cell transplantation. MOLECULAR AND CELLULAR THERAPIES 2014; 2:19. [PMID: 26056586 PMCID: PMC4452047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 06/05/2014] [Indexed: 11/21/2023]
Abstract
Progress is being made in developing neuroprotective strategies for traumatic brain injuries; however, there will never be a therapy that will fully preserve neurons that are injured from moderate to severe head injuries. Therefore, to restore neurological function, regenerative strategies will be required. Given the limited regenerative capacity of the resident neural precursors of the CNS, many investigators have evaluated the regenerative potential of transplanted precursors. Unfortunately, these precursors do not thrive when engrafted without a biomaterial scaffold. In this article we review the types of natural and synthetic materials that are being used in brain tissue engineering applications for traumatic brain injury and stroke. We also analyze modifications of the scaffolds including immobilizing drugs, growth factors and extracellular matrix molecules to improve CNS regeneration and functional recovery. We conclude with a discussion of some of the challenges that remain to be solved towards repairing and regenerating the brain.
Collapse
Affiliation(s)
- Nolan B Skop
- />Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA
- />Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102 USA
| | - Frances Calderon
- />Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA
| | - Cheul H Cho
- />Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102 USA
| | - Chirag D Gandhi
- />Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA
- />Department of Neurological Surgery, Rutgers University-New Jersey Medical School, New Jersey Medical School, Newark, NJ 07103 USA
| | - Steven W Levison
- />Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA
| |
Collapse
|
35
|
Gold EM, Su D, López-Velázquez L, Haus DL, Perez H, Lacuesta GA, Anderson AJ, Cummings BJ. Functional assessment of long-term deficits in rodent models of traumatic brain injury. Regen Med 2014; 8:483-516. [PMID: 23826701 DOI: 10.2217/rme.13.41] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Traumatic brain injury (TBI) ranks as the leading cause of mortality and disability in the young population worldwide. The annual US incidence of TBI in the general population is estimated at 1.7 million per year, with an estimated financial burden in excess of US$75 billion a year in the USA alone. Despite the prevalence and cost of TBI to individuals and society, no treatments have passed clinical trial to clinical implementation. The rapid expansion of stem cell research and technology offers an alternative to traditional pharmacological approaches targeting acute neuroprotection. However, preclinical testing of these approaches depends on the selection and characterization of appropriate animal models. In this article we consider the underlying pathophysiology for the focal and diffuse TBI subtypes, discuss the existing preclinical TBI models and functional outcome tasks used for assessment of injury and recovery, identify criteria particular to preclinical animal models of TBI in which stem cell therapies can be tested for safety and efficacy, and review these criteria in the context of the existing TBI literature. We suggest that 2 months post-TBI is the minimum period needed to evaluate human cell transplant efficacy and safety. Comprehensive review of the published TBI literature revealed that only 32% of rodent TBI papers evaluated functional outcome ≥1 month post-TBI, and only 10% evaluated functional outcomes ≥2 months post-TBI. Not all published papers that evaluated functional deficits at a minimum of 2 months post-TBI reported deficits; hence, only 8.6% of overall TBI papers captured in this review demonstrated functional deficits at 2 months or more postinjury. A 2-month survival and assessment period would allow sufficient time for differentiation and integration of human neural stem cells with the host. Critically, while trophic effects might be observed at earlier time points, it will also be important to demonstrate the sustainability of such an effect, supporting the importance of an extended period of in vivo observation. Furthermore, regulatory bodies will likely require at least 6 months survival post-transplantation for assessment of toxicology/safety, particularly in the context of assessing cell abnormalities.
Collapse
Affiliation(s)
- Eric M Gold
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine 2030 Gross Hall, CA 92697-1705, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Mahmood A, Wu H, Qu C, Mahmood S, Xiong Y, Kaplan DL, Chopp M. Suppression of neurocan and enhancement of axonal density in rats after treatment of traumatic brain injury with scaffolds impregnated with bone marrow stromal cells. J Neurosurg 2014; 120:1147-55. [PMID: 24460490 DOI: 10.3171/2013.12.jns131362] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECT Neurocan is a major form of growth-inhibitory molecule (growth-IM) that suppresses axonal regeneration after neural injury. Bone marrow stromal cells (MSCs) have been shown to inhibit neurocan expression in vitro and in animal models of cerebral ischemia. Therefore, the present study was designed to investigate the effects of treatment of MSCs impregnated with collagen scaffolds on neurocan expression after traumatic brain injury (TBI). METHODS Adult male Wistar rats were injured with controlled cortical impact and treated with saline, human MSCs (hMSCs) (3 × 10(6)) alone, or hMSCs (3 × 10(6)) impregnated into collagen scaffolds (scaffold + hMSCs) transplanted into the lesion cavity 7 days after TBI (20 rats per group). Rats were sacrificed 14 days after TBI, and brain tissues were harvested for immunohistochemical studies, Western blot analyses, laser capture microdissections, and quantitative real-time reverse transcriptase polymerase chain reaction (qRT-PCR) to evaluate neurocan protein and gene expressions after various treatments. RESULTS Animals treated with scaffold + hMSCs after TBI showed increased axonal and synaptic densities compared with the other groups. Scaffold + hMSC treatment was associated with reduced TBI-induced neurocan protein expression and upregulated growth-associated protein 43 (GAP-43) and synaptophysin expression in the lesion boundary zone. In addition, animals in the scaffold + hMSC group had decreased neurocan transcription in reactive astrocytes after TBI. Reduction of neurocan expression was significantly greater in the scaffold + hMSC group than in the group treated with hMSCs alone. CONCLUSIONS The results of this study show that transplanting hMSCs with scaffolds enhances the effect of hMSCs on axonal plasticity in TBI rats. This enhanced axonal plasticity may partially be attributed to the downregulation of neurocan expression by hMSC treatment after injury.
Collapse
|
37
|
Moore TL, Pessina MA, Finklestein SP, Kramer BC, Killiany RJ, Rosene DL. Recovery of fine motor performance after ischemic damage to motor cortex is facilitated by cell therapy in the rhesus monkey. Somatosens Mot Res 2013; 30:185-96. [PMID: 23758412 PMCID: PMC6503838 DOI: 10.3109/08990220.2013.790806] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We investigated the efficacy on recovery of function following controlled cortical ischemia in the monkey of the investigational cell drug product, CNTO 0007. This drug contains a cellular component, human umbilical tissue-derived cells, in a proprietary thaw and inject formulation. Results demonstrate significantly better recovery of motor function in the treatment group with no difference between groups in the volume or surface area of ischemic damage, suggesting that the cells stimulated plasticity.
Collapse
Affiliation(s)
- Tara L Moore
- Department of Anatomy & Neurobiology, Boston University School of Medicine , Boston, MA , USA
| | | | | | | | | | | |
Collapse
|
38
|
Mahmood A, Wu H, Qu C, Mahmood S, Xiong Y, Kaplan D, Chopp M. Down-regulation of Nogo-A by collagen scaffolds impregnated with bone marrow stromal cell treatment after traumatic brain injury promotes axonal regeneration in rats. Brain Res 2013; 1542:41-8. [PMID: 24177046 DOI: 10.1016/j.brainres.2013.10.045] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 10/23/2013] [Indexed: 10/26/2022]
Abstract
Nogo-A is a major form of growth inhibitory molecule (growth-IM) which inhibits axonal regeneration and neurite regrowth after neural injury. Bone marrow stromal cells (MSCs) have been shown to inhibit Nogo-A expression in vitro and in cerebral ischemic animal models. The present study was designed to investigate the effects of treatment with human MSCs (hMSCs) impregnated into collagen scaffolds on the expression of Nogo-A and axonal plasticity after traumatic brain injury (TBI). Adult male Wistar rats were injured with controlled cortical impact and treated either with saline, hMSCs-alone or hMSCs impregnated into collagen scaffolds (scaffold+hMSC) transplanted into the lesion cavity 7 days after TBI. Rats were sacrificed 14 days after TBI and brain tissues were harvested for immunohistochemical studies, Western blot analysis, laser capture microdissections and qRT-PCR to evaluate axonal density and Nogo-A protein and gene expressions. Our data showed that treatment of TBI with scaffold+hMSC significantly decreased TBI-induced Nogo-A protein expression and increased axonal density compared to saline and hMSC-alone treatments. In addition, scaffold+hMSC transplantation decreased Nogo-A transcription in oligodendrocytes after TBI. Scaffold+hMSC treatment was superior to hMSC-alone treatment in suppressing Nogo-A expression and enhancing axonal regeneration after TBI. Our data suggest that transplanting hMSCs with scaffolds down-regulates Nogo-A transcription and protein expression which may partially contribute to the enhanced axonal regeneration after TBI.
Collapse
Affiliation(s)
- Asim Mahmood
- Department of Neurosurgery, 2799W Grand Blvd, Henry Ford Hospital, Detroit, MI 48202, USA.
| | - Hongtao Wu
- Department of Neurosurgery, 2799W Grand Blvd, Henry Ford Hospital, Detroit, MI 48202, USA.
| | - Changsheng Qu
- Department of Neurosurgery, 2799W Grand Blvd, Henry Ford Hospital, Detroit, MI 48202, USA.
| | - Selina Mahmood
- Department of Neurosurgery, 2799W Grand Blvd, Henry Ford Hospital, Detroit, MI 48202, USA.
| | - Ye Xiong
- Department of Neurosurgery, 2799W Grand Blvd, Henry Ford Hospital, Detroit, MI 48202, USA.
| | - David Kaplan
- Department of Biomedical Engineering, Science and Technology Center, Room 251, Tufts University, Boston, MA 02155, USA.
| | - Michael Chopp
- Department of Neurology, 2799W Grand Blvd, Henry Ford Hospital, Detroit, MI 48202, USA; Department of Physics, Oakland University, 2200 North Squirrel Road, Rochester, MI 48309-4401, USA.
| |
Collapse
|
39
|
Castillo-Melendez M, Yawno T, Jenkin G, Miller SL. Stem cell therapy to protect and repair the developing brain: a review of mechanisms of action of cord blood and amnion epithelial derived cells. Front Neurosci 2013; 7:194. [PMID: 24167471 PMCID: PMC3807037 DOI: 10.3389/fnins.2013.00194] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 10/07/2013] [Indexed: 12/12/2022] Open
Abstract
In the research, clinical, and wider community there is great interest in the use of stem cells to reduce the progression, or indeed repair brain injury. Perinatal brain injury may result from acute or chronic insults sustained during fetal development, during the process of birth, or in the newborn period. The most readily identifiable outcome of perinatal brain injury is cerebral palsy, however, this is just one consequence in a spectrum of mild to severe neurological deficits. As we review, there are now clinical trials taking place worldwide targeting cerebral palsy with stem cell therapies. It will likely be many years before strong evidence-based results emerge from these trials. With such trials underway, it is both appropriate and timely to address the physiological basis for the efficacy of stem-like cells in preventing damage to, or regenerating, the newborn brain. Appropriate experimental animal models are best placed to deliver this information. Cell availability, the potential for immunological rejection, ethical, and logistical considerations, together with the propensity for native cells to form teratomas, make it unlikely that embryonic or fetal stem cells will be practical. Fortunately, these issues do not pertain to the use of human amnion epithelial cells (hAECs), or umbilical cord blood (UCB) stem cells that are readily and economically obtained from the placenta and umbilical cord discarded at birth. These cells have the potential for transplantation to the newborn where brain injury is diagnosed or even suspected. We will explore the novel characteristics of hAECs and undifferentiated UCB cells, as well as UCB-derived endothelial progenitor cells (EPCs) and mesenchymal stem cells (MSCs), and how immunomodulation and anti-inflammatory properties are principal mechanisms of action that are common to these cells, and which in turn may ameliorate the cerebral hypoxia and inflammation that are final pathways in the pathogenesis of perinatal brain injury.
Collapse
Affiliation(s)
- Margie Castillo-Melendez
- The Ritchie Centre, Monash Institute of Medical Research, Monash University Clayton, VIC, Australia
| | | | | | | |
Collapse
|
40
|
Lin P, Correa D, Kean TJ, Awadallah A, Dennis JE, Caplan AI. Serial transplantation and long-term engraftment of intra-arterially delivered clonally derived mesenchymal stem cells to injured bone marrow. Mol Ther 2013; 22:160-8. [PMID: 24067545 DOI: 10.1038/mt.2013.221] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 09/15/2013] [Indexed: 12/24/2022] Open
Abstract
It has been hypothesized that mesenchymal stem cells (MSCs) home to sites of injury. Nevertheless, efficient delivery of MSCs to target organs and description of their ultimate fate remain major challenges. We provide evidence that intra-arterially (IA) injected MSCs selectively engraft from the circulation as perivascular cells in the bone marrow (BM) after a localized radiation injury. Luciferase-expressing MSCs, derived from a conditionally immortalized clone (BMC-9) representing a pure population of cells, were arterially delivered into mice irradiated in one leg. Cell distribution was measured by bioluminescent imaging and final destination assessed by luciferase immunolocalization. IA injections resulted in engraftment only in the irradiated leg where cells localize and proliferate abluminal to the BM vasculature, a phenomenon not replicated with intravenous injections or with IA injections of kidney cells harvested from the same donor used for MSCs. Furthermore, MSCs harvested from the engrafted marrow and serially transplanted retain the ability to selectively engraft at sites of injury. This study demonstrates that MSCs can serially engraft at sites of injury from the circulation, that they reside in the perivascular space, and that arterial delivery is more efficient than venous delivery for cell engraftment.
Collapse
Affiliation(s)
- Paul Lin
- 1] Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA [2] Skeletal Research Center, Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Diego Correa
- Skeletal Research Center, Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Thomas J Kean
- Benaroya Research Institute, Seattle, Washington, USA
| | - Amad Awadallah
- Skeletal Research Center, Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Arnold I Caplan
- Skeletal Research Center, Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
41
|
da Cruz e Alves-de-Moraes LB, Ribeiro-Paes JT, Longo BM, Ferrazoli EG, de Andrade TGCS. Effect of the bone marrow cell transplantation on elevated plus-maze performance in hippocampal-injured mice. Behav Brain Res 2013; 248:32-40. [PMID: 23578758 DOI: 10.1016/j.bbr.2013.03.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 03/25/2013] [Accepted: 03/31/2013] [Indexed: 11/15/2022]
Abstract
Several reports have shown that the hippocampus plays an important role in different aspects of the emotional control. There is evidence that lesions in this structure cause behavioral disinhibition, with reduction of reactions expressing fear and anxiety. Thus, to portray the aptitude of cell therapy to abrogate injuries of hippocampal tissue, we examined the behavioral effects of bone marrow mononuclear cells (BMMCs) transplantation on C57BL/6 mice that had the hippocampus damaged by electrolytic lesion. For this purpose, mice received, seven days after bilateral electrolytic lesion in the dorsal hippocampus, culture medium or BMMCs expressing the enhanced green fluorescent protein (EGFP) transgene. One week after transplantation, animals were tested in the elevated plus-maze (EPM). On the whole, three assessment sessions in the EPM were carried out, with seven days separating each trial. Thirty-five days after the induction of injury, mice were sacrificed and their brains removed for immunohistochemistry. The behavioral evaluation showed that the hippocampal lesion caused disinhibition, an effect which was slightly lessened, from the second EPM test, in transplanted subjects. On the other hand, immunohistochemical data revealed an insignificant presence of EGFP(+) cells inside the brains of injured mice. In view of such scenario, we hypothesized that the subtle rehabilitation of the altered behavior might be a result from a paracrine effect from the transplanted cells. This might have been caused by the release of bioactive factors capable of boosting endogenous recuperative mechanisms for a partial regaining of the hippocampal functions.
Collapse
|
42
|
Falavigna A, da Costa JC. Mesenchymal autologous stem cells. World Neurosurg 2013; 83:236-50. [PMID: 23402865 DOI: 10.1016/j.wneu.2013.02.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 01/24/2013] [Accepted: 02/05/2013] [Indexed: 02/07/2023]
Abstract
The use of cell-based therapies for spinal cord injuries has recently gained prominence as a potential therapy or component of a combination strategy. Experimental and clinical studies have been performed using mesenchymal stem cell therapy to treat spinal cord injuries with encouraging results. However, there have been reports on the adverse effects of these stem cell-based therapies, especially in the context of tumor modulation. This article surveys the literature relevant to the potential of mesenchymal autologous stem cells for spinal cord injuries and their clinical implications.
Collapse
Affiliation(s)
- Asdrubal Falavigna
- Department of Neurosurgery, Medical School of the University of Caxias do Sul, Caxias do Sul, Brazil.
| | - Jaderson Costa da Costa
- Neurology Service and Instituto do Cérebro, Pontifical Catholic University of Rio Grande do Sul, Brazil
| |
Collapse
|
43
|
Kholodenko IV, Konieva AA, Kholodenko RV, Yarygin KN. Molecular mechanisms of migration and homing of intravenously transplanted mesenchymal stem cells. ACTA ACUST UNITED AC 2013. [DOI: 10.7243/2050-1218-2-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
44
|
Mahmood A, Wu H, Qu C, Xiong Y, Chopp M. Effects of treating traumatic brain injury with collagen scaffolds and human bone marrow stromal cells on sprouting of corticospinal tract axons into the denervated side of the spinal cord. J Neurosurg 2012. [PMID: 23198801 DOI: 10.3171/2012.11.jns12753] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT This study was designed to investigate how transplantation into injured brain of human bone marrow stromal cells (hMSCs) impregnated in collagen scaffolds affects axonal sprouting in the spinal cord after traumatic brain injury (TBI) in rats. Also investigated was the relationship of axonal sprouting to sensorimotor functional recovery after treatment. METHODS Adult male Wistar rats (n = 24) underwent a controlled cortical impact injury and were divided into three equal groups (8 rats/group). The two treatment groups received either hMSCs (3 × 10(6)) alone or hMSC (3 × 10(6))-impregnated collagen scaffolds transplanted into the lesion cavity. In the control group, saline was injected into the lesion cavity. All treatments were performed 7 days after TBI. On Day 21 after TBI, a 10% solution of biotinylated dextran amine (10,000 MW) was stereotactically injected into the contralateral motor cortex to label the corticospinal tract (CST) originating from this area. Sensorimotor function was tested using the modified neurological severity score (mNSS) and foot-fault tests performed on Days 1, 7, 14, 21, 28, and 35 after TBI. Spatial learning was tested with Morris water maze test on Days 31-35 after TBI. All rats were sacrificed on Day 35 after TBI, and brain and spinal cord (cervical and lumbar) sections were stained immunohistochemically for histological analysis. RESULTS Few biotinylated dextran amine-labeled CST fibers crossing over the midline were found in the contralateral spinal cord transverse sections at both cervical and lumbar levels in saline-treated (control) rats. However, hMSC-alone treatment significantly increased axonal sprouting from the intact CST into the denervated side of the gray matter of both cervical and lumbar levels of the spinal cord (p < 0.05). Also, this axonal sprouting was significantly more in the scaffold+hMSC group compared with the hMSC-alone group (p < 0.05). Sensorimotor functional analysis showed significant improvement of mNSS (p < 0.05) and foot-fault tests (p < 0.05) in hMSC-alone and scaffold+hMSC-treated rats compared with controls (p < 0.05). Functional improvement, however, was significantly greater in the scaffold+hMSC group compared with the hMSC-alone group (p < 0.05). Morris water maze testing also showed significant improvement in spatial learning in scaffold+hMSC and hMSC-alone groups compared with the control group (p < 0.05), with rats in the scaffold+hMSC group performing significantly better than those in the hMSC-alone group (p < 0.05). Pearson correlation data showed significant correlation between the number of crossing CST fibers detected and sensorimotor recovery (p < 0.05). CONCLUSIONS Axonal plasticity plays an important role in neurorestoration after TBI. Transplanting hMSCs with scaffolds enhances the effect of hMSCs on axonal sprouting of CST fibers from the contralateral intact cortex into the denervated side of spinal cord after TBI. This enhanced axonal regeneration may at least partially contribute to the therapeutic benefits of treating TBI with hMSCs.
Collapse
Affiliation(s)
- Asim Mahmood
- Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan 48202, USA.
| | | | | | | | | |
Collapse
|
45
|
Kemp K, Gray E, Wilkins A, Scolding N. Purkinje cell fusion and binucleate heterokaryon formation in multiple sclerosis cerebellum. Brain 2012; 135:2962-72. [PMID: 22975392 DOI: 10.1093/brain/aws226] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
A major conceptual consideration in both endogenous and therapeutic central nervous system repair is how damaged (or senescent) neurons, given their often enormously complex and extensive network of connections, can possibly be replaced. The recent observation of fusion of circulating bone marrow cells with, in particular, cerebellar Purkinje cells, as well as the subsequent formation of stable heterokaryons, offers a tantalizing potential solution to this difficulty. Here, we have explored Purkinje cell fusion and heterokaryon formation in the human brain and the influence of central nervous system inflammation. We analysed post-mortem cerebellum tissue from patients who had multiple sclerosis and from appropriate controls. Purkinje cells were analysed for heterokaryon formation using immunohistochemistry techniques and chromosome composition using fluorescence in situ hybridization. For the first time in humans we show a disease-related increase in Purkinje cell fusion and heterokaryon formation. We have shown that heterokaryon formation takes place in control subjects, and that the frequency of this event is considerably increased in patients with multiple sclerosis, the prototypical inflammatory brain disease, with ~0.4% of Purkinje cells being binucleate heterokaryons. No mononucleate polyploid Purkinje cell heterokaryons were found. The observation that heterokaryon formation in the cerebellum occurs as part of the central nervous system inflammatory reaction suggests a potential mechanism of neural repair. It also suggests an exciting new avenue for therapeutic intervention, as enhancement or manipulation of fusion events may have a therapeutic role in cellular protection in multiple sclerosis.
Collapse
Affiliation(s)
- Kevin Kemp
- Multiple Sclerosis and Stem Cell, School of Clinical Sciences, University of Bristol, Bristol, BS16 1JB, UK.
| | | | | | | |
Collapse
|
46
|
Journey of mesenchymal stem cells for homing: strategies to enhance efficacy and safety of stem cell therapy. Stem Cells Int 2012; 2012:342968. [PMID: 22754575 PMCID: PMC3382267 DOI: 10.1155/2012/342968] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 04/06/2012] [Accepted: 04/17/2012] [Indexed: 12/11/2022] Open
Abstract
Human mesenchymal stem cells (MSCs) communicate with other cells in the human body and appear to "home" to areas of injury in response to signals of cellular damage, known as homing signals. This review of the state of current research on homing of MSCs suggests that favorable cellular conditions and the in vivo environment facilitate and are required for the migration of MSCs to the site of insult or injury in vivo. We review the current understanding of MSC migration and discuss strategies for enhancing both the environmental and cellular conditions that give rise to effective homing of MSCs. This may allow MSCs to quickly find and migrate to injured tissues, where they may best exert clinical benefits resulting from improved homing and the presence of increased numbers of MSCs.
Collapse
|
47
|
Bonilla C, Zurita M, Otero L, Aguayo C, Rico MA, Rodríguez A, Vaquero J. Failure of Delayed Intravenous Administration of Bone Marrow Stromal Cells after Traumatic Brain Injury. J Neurotrauma 2012; 29:394-400. [DOI: 10.1089/neu.2011.2101] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Affiliation(s)
- Celia Bonilla
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Mercedes Zurita
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Laura Otero
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Concepción Aguayo
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Miguel A. Rico
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Alicia Rodríguez
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Jesús Vaquero
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
- Service of Neurosurgery, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| |
Collapse
|
48
|
Anderson AJ, Haus DL, Hooshmand MJ, Perez H, Sontag CJ, Cummings BJ. Achieving stable human stem cell engraftment and survival in the CNS: is the future of regenerative medicine immunodeficient? Regen Med 2011; 6:367-406. [PMID: 21548741 DOI: 10.2217/rme.11.22] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
There is potential for a variety of stem cell populations to mediate repair in the diseased or injured CNS; in some cases, this theoretical possibility has already transitioned to clinical safety testing. However, careful consideration of preclinical animal models is essential to provide an appropriate assessment of stem cell safety and efficacy, as well as the basic biological mechanisms of stem cell action. This article examines the lessons learned from early tissue, organ and hematopoietic grafting, the early assumptions of the stem cell and CNS fields with regard to immunoprivilege, and the history of success in stem cell transplantation into the CNS. Finally, we discuss strategies in the selection of animal models to maximize the predictive validity of preclinical safety and efficacy studies.
Collapse
Affiliation(s)
- Aileen J Anderson
- Sue & Bill Gross Stem Cell Center, 845 Health Science Road, UC Irvine, Irvine, CA 92697-1705, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Kemp K, Mallam E, Hares K, Witherick J, Scolding N, Wilkins A. Mesenchymal stem cells restore frataxin expression and increase hydrogen peroxide scavenging enzymes in Friedreich ataxia fibroblasts. PLoS One 2011; 6:e26098. [PMID: 22016819 PMCID: PMC3189234 DOI: 10.1371/journal.pone.0026098] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 09/19/2011] [Indexed: 01/01/2023] Open
Abstract
Dramatic advances in recent decades in understanding the genetics of Friedreich ataxia (FRDA)--a GAA triplet expansion causing greatly reduced expression of the mitochondrial protein frataxin--have thus far yielded no therapeutic dividend, since there remain no effective treatments that prevent or even slow the inevitable progressive disability in affected individuals. Clinical interventions that restore frataxin expression are attractive therapeutic approaches, as, in theory, it may be possible to re-establish normal function in frataxin deficient cells if frataxin levels are increased above a specific threshold. With this in mind several drugs and cytokines have been tested for their ability to increase frataxin levels. Cell transplantation strategies may provide an alternative approach to this therapeutic aim, and may also offer more widespread cellular protective roles in FRDA. Here we show a direct link between frataxin expression in fibroblasts derived from FRDA patients with both decreased expression of hydrogen peroxide scavenging enzymes and increased sensitivity to hydrogen peroxide-mediated toxicity. We demonstrate that normal human mesenchymal stem cells (MSCs) induce both an increase in frataxin gene and protein expression in FRDA fibroblasts via secretion of soluble factors. Finally, we show that exposure to factors produced by human MSCs increases resistance to hydrogen peroxide-mediated toxicity in FRDA fibroblasts through, at least in part, restoring the expression of the hydrogen peroxide scavenging enzymes catalase and glutathione peroxidase 1. These findings suggest, for the first time, that stem cells may increase frataxin levels in FRDA and transplantation of MSCs may offer an effective treatment for these patients.
Collapse
Affiliation(s)
- Kevin Kemp
- Multiple Sclerosis and Stem Cell Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom.
| | | | | | | | | | | |
Collapse
|
50
|
Azari MF, Mathias L, Ozturk E, Cram DS, Boyd RL, Petratos S. Mesenchymal stem cells for treatment of CNS injury. Curr Neuropharmacol 2011; 8:316-23. [PMID: 21629440 PMCID: PMC3080589 DOI: 10.2174/157015910793358204] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Revised: 05/15/2010] [Accepted: 05/25/2010] [Indexed: 02/06/2023] Open
Abstract
Brain and spinal cord injuries present significant therapeutic challenges. The treatments available for these conditions are largely ineffective, partly due to limitations in directly targeting the therapeutic agents to sites of pathology within the central nervous system (CNS). The use of stem cells to treat these conditions presents a novel therapeutic strategy. A variety of stem cell treatments have been examined in animal models of CNS trauma. Many of these studies have used stem cells as a cell-replacement strategy. These investigations have also highlighted the significant limitations of this approach. Another potential strategy for stem cell therapy utilises stem cells as a delivery mechanism for therapeutic molecules. This review surveys the literature relevant to the potential of mesenchymal stem cells for delivery of therapeutic agents in CNS trauma in humans.
Collapse
Affiliation(s)
- Michael F Azari
- Monash Immunology and Stem Cell Laboratories, School of Biomedical Sciences, Faculty of Medicine, Monash University, Clayton, Vic. Australia
| | | | | | | | | | | |
Collapse
|