1
|
Schantz SL, Sneed SE, Fagan MM, Golan ME, Cheek SR, Kinder HA, Duberstein KJ, Kaiser EE, West FD. Human-Induced Pluripotent Stem Cell-Derived Neural Stem Cell Therapy Limits Tissue Damage and Promotes Tissue Regeneration and Functional Recovery in a Pediatric Piglet Traumatic-Brain-Injury Model. Biomedicines 2024; 12:1663. [PMID: 39200128 PMCID: PMC11351842 DOI: 10.3390/biomedicines12081663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/12/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in pediatric patients and often results in delayed neural development and altered connectivity, leading to lifelong learning, memory, behavior, and motor function deficits. Induced pluripotent stem cell-derived neural stem cells (iNSCs) may serve as a novel multimodal therapeutic as iNSCs possess neuroprotective, regenerative, and cell-replacement capabilities post-TBI. In this study, we evaluated the effects of iNSC treatment on cellular, tissue, and functional recovery in a translational controlled cortical impact TBI piglet model. Five days post-craniectomy (n = 6) or TBI (n = 18), iNSCs (n = 7) or PBS (n = 11) were injected into perilesional brain tissue. Modified Rankin Scale (mRS) neurological evaluation, magnetic resonance imaging, and immunohistochemistry were performed over the 12-week study period. At 12-weeks post-transplantation, iNSCs showed long-term engraftment and differentiation into neurons, astrocytes, and oligodendrocytes. iNSC treatment enhanced endogenous neuroprotective and regenerative activities indicated by decreasing intracerebral immune responses, preserving endogenous neurons, and increasing neuroblast formation. These cellular changes corresponded with decreased hemispheric atrophy, midline shift, and lesion volume as well as the preservation of cerebral blood flow. iNSC treatment increased piglet survival and decreased mRS scores. The results of this study in a predictive pediatric large-animal pig model demonstrate that iNSC treatment is a robust multimodal therapeutic that has significant promise in potentially treating human pediatric TBI patients.
Collapse
Affiliation(s)
- Sarah L. Schantz
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA; (S.L.S.); (S.E.S.); (M.E.G.); (S.R.C.); (H.A.K.); (K.J.D.)
- Biomedical and Health Sciences Institute, University of Georgia, Athens, GA 30602, USA
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
| | - Sydney E. Sneed
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA; (S.L.S.); (S.E.S.); (M.E.G.); (S.R.C.); (H.A.K.); (K.J.D.)
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
| | - Madison M. Fagan
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA; (S.L.S.); (S.E.S.); (M.E.G.); (S.R.C.); (H.A.K.); (K.J.D.)
- Biomedical and Health Sciences Institute, University of Georgia, Athens, GA 30602, USA
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
| | - Morgane E. Golan
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA; (S.L.S.); (S.E.S.); (M.E.G.); (S.R.C.); (H.A.K.); (K.J.D.)
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
| | - Savannah R. Cheek
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA; (S.L.S.); (S.E.S.); (M.E.G.); (S.R.C.); (H.A.K.); (K.J.D.)
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
| | - Holly A. Kinder
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA; (S.L.S.); (S.E.S.); (M.E.G.); (S.R.C.); (H.A.K.); (K.J.D.)
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
| | - Kylee J. Duberstein
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA; (S.L.S.); (S.E.S.); (M.E.G.); (S.R.C.); (H.A.K.); (K.J.D.)
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
| | - Erin E. Kaiser
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA; (S.L.S.); (S.E.S.); (M.E.G.); (S.R.C.); (H.A.K.); (K.J.D.)
- Biomedical and Health Sciences Institute, University of Georgia, Athens, GA 30602, USA
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
| | - Franklin D. West
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA; (S.L.S.); (S.E.S.); (M.E.G.); (S.R.C.); (H.A.K.); (K.J.D.)
- Biomedical and Health Sciences Institute, University of Georgia, Athens, GA 30602, USA
- Animal and Dairy Science Department, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
2
|
Davis CK, Arruri V, Joshi P, Vemuganti R. Non-pharmacological interventions for traumatic brain injury. J Cereb Blood Flow Metab 2024; 44:641-659. [PMID: 38388365 PMCID: PMC11197135 DOI: 10.1177/0271678x241234770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/24/2024]
Abstract
Heterogeneity and variability of symptoms due to the type, site, age, sex, and severity of injury make each case of traumatic brain injury (TBI) unique. Considering this, a universal treatment strategy may not be fruitful in managing outcomes after TBI. Most of the pharmacological therapies for TBI aim at modifying a particular pathway or molecular process in the sequelae of secondary injury rather than a holistic approach. On the other hand, non-pharmacological interventions such as hypothermia, hyperbaric oxygen, preconditioning with dietary adaptations, exercise, environmental enrichment, deep brain stimulation, decompressive craniectomy, probiotic use, gene therapy, music therapy, and stem cell therapy can promote healing by modulating multiple neuroprotective mechanisms. In this review, we discussed the major non-pharmacological interventions that are being tested in animal models of TBI as well as in clinical trials. We evaluated the functional outcomes of various interventions with an emphasis on the links between molecular mechanisms and outcomes after TBI.
Collapse
Affiliation(s)
- Charles K Davis
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Vijay Arruri
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Pallavi Joshi
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| |
Collapse
|
3
|
Friedel J, Pierre S, Kolbinger A, Schäufele TJ, Aliraj B, Weigert A, Scholich K. Mast cell-derived interleukin-4 mediates activation of dendritic cell during toll-like receptor 2-mediated inflammation. Front Immunol 2024; 15:1353922. [PMID: 38745645 PMCID: PMC11091258 DOI: 10.3389/fimmu.2024.1353922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/17/2024] [Indexed: 05/16/2024] Open
Abstract
Introduction During an innate inflammation, immune cells form distinct pro- and anti-inflammatory regions around pathogen-containing core-regions. Mast cells are localized in an anti-inflammatory microenvironment during the resolution of an innate inflammation, suggesting antiinflammatory roles of these cells. Methods High-content imaging was used to investigated mast cell-dependent changes in the regional distribution of immune cells during an inflammation, induced by the toll-like receptor (TLR)-2 agonist zymosan. Results The distance between the zymosan-containing core-region and the anti-inflammatory region, described by M2-like macrophages, increased in mast cell-deficient mice. Absence of mast cells abolished dendritic cell (DC) activation, as determined by CD86-expression and localized the DCs in greater distance to zymosan particles. The CD86- DCs had a higher expression of the pro-inflammatory interleukins (IL)-1β and IL-12/23p40 as compared to activated CD86+ DCs. IL-4 administration restored CD86 expression, cytokine expression profile and localization of the DCs in mast cell-deficient mice. The IL-4 effects were mast cell-specific, since IL-4 reduction by eosinophil depletion did not affect activation of DCs. Discussion We found that mast cells induce DC activation selectively at the site of inflammation and thereby determine their localization within the inflammation. Overall, mast cells have antiinflammatory functions in this inflammation model and limit the size of the pro-inflammatory region surrounding the zymosan-containing core region.
Collapse
Affiliation(s)
- Joschua Friedel
- Institute of Clinical Pharmacology, Goethe University, Frankfurt, Germany
| | - Sandra Pierre
- Institute of Clinical Pharmacology, Goethe University, Frankfurt, Germany
| | - Anja Kolbinger
- Institute of Clinical Pharmacology, Goethe University, Frankfurt, Germany
| | - Tim J. Schäufele
- Institute of Clinical Pharmacology, Goethe University, Frankfurt, Germany
| | - Blerina Aliraj
- Institute of Biochemistry I, Faculty of Medicine, Goethe University, Frankfurt, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe University, Frankfurt, Germany
| | - Klaus Scholich
- Institute of Clinical Pharmacology, Goethe University, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases CIMD, Frankfurt, Germany
| |
Collapse
|
4
|
Syzdykbayev M, Kazymov M, Aubakirov M, Kurmangazina A, Kairkhanov E, Kazangapov R, Bryzhakhina Z, Imangazinova S, Sheinin A. A Modern Approach to the Treatment of Traumatic Brain Injury. MEDICINES (BASEL, SWITZERLAND) 2024; 11:10. [PMID: 38786549 PMCID: PMC11123131 DOI: 10.3390/medicines11050010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/18/2024] [Accepted: 04/27/2024] [Indexed: 05/25/2024]
Abstract
Background: Traumatic brain injury manifests itself in various forms, ranging from mild impairment of consciousness to severe coma and death. Traumatic brain injury remains one of the leading causes of morbidity and mortality. Currently, there is no therapy to reverse the effects associated with traumatic brain injury. New neuroprotective treatments for severe traumatic brain injury have not achieved significant clinical success. Methods: A literature review was performed to summarize the recent interdisciplinary findings on management of traumatic brain injury from both clinical and experimental perspective. Results: In the present review, we discuss the concepts of traditional and new approaches to treatment of traumatic brain injury. The recent development of different drug delivery approaches to the central nervous system is also discussed. Conclusions: The management of traumatic brain injury could be aimed either at the pathological mechanisms initiating the secondary brain injury or alleviating the symptoms accompanying the injury. In many cases, however, the treatment should be complex and include a variety of medical interventions and combination therapy.
Collapse
Affiliation(s)
- Marat Syzdykbayev
- Department of Hospital Surgery, Anesthesiology and Reanimatology, Semey Medical University, Semey 071400, Kazakhstan
| | - Maksut Kazymov
- Department of General Practitioners, Semey Medical University, Semey 071400, Kazakhstan
| | - Marat Aubakirov
- Department of Pediatric Surgery, Semey Medical University, Semey 071400, Kazakhstan
| | - Aigul Kurmangazina
- Committee for Medical and Pharmaceutical Control of the Ministry of Health of the Republic of Kazakhstan for East Kazakhstan Region, Ust-Kamenogorsk 070004, Kazakhstan
| | - Ernar Kairkhanov
- Pavlodar Branch of Semey Medical University, Pavlodar S03Y3M1, Kazakhstan
| | - Rustem Kazangapov
- Pavlodar Branch of Semey Medical University, Pavlodar S03Y3M1, Kazakhstan
| | - Zhanna Bryzhakhina
- Department Psychiatry and Narcology, Semey Medical University, Semey 071400, Kazakhstan
| | - Saule Imangazinova
- Department of Therapy, Astana Medical University, Astana 010000, Kazakhstan
| | - Anton Sheinin
- Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv 69978, Israel
| |
Collapse
|
5
|
Liu S, Shi L, Huang T, Luo Y, Chen Y, Li S, Wang Z. Neural Stem Cells Transplanted into Rhesus Monkey Cortical Traumatic Brain Injury Can Survive and Differentiate into Neurons. Int J Mol Sci 2024; 25:1642. [PMID: 38338922 PMCID: PMC10855641 DOI: 10.3390/ijms25031642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Cortical traumatic brain injury (TBI) is a major cause of cognitive impairment accompanied by motor and behavioral deficits, and there is no effective treatment strategy in the clinic. Cell transplantation is a promising therapeutic strategy, and it is necessary to verify the survival and differentiation of cells after transplantation in large animal models like rhesus monkeys. In this study, we transplanted neural stem cells (NSCs) and simultaneously injected basic fibroblast growth factor/epidermal growth factor (bFGF/EGF) into the cortex (visual and sensory cortices) of rhesus monkeys with superficial TBI. The results showed that the transplanted NSCs did not enter the cerebrospinal fluid (CSF) and were confined to the transplantation site for at least one year. The transplanted NSCs differentiated into mature neurons that formed synaptic connections with host neurons, but glial scar formation between the graft and the host tissue did not occur. This study is the first to explore the repairing effect of transplanting NSCs into the superficial cerebral cortex of rhesus monkeys after TBI, and the results show the ability of NSCs to survive long-term and differentiate into neurons, demonstrating the potential of NSC transplantation for cortical TBI.
Collapse
Affiliation(s)
- Shuyi Liu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China; (S.L.); (L.S.); (T.H.); (Y.L.); (S.L.)
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| | - Liping Shi
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China; (S.L.); (L.S.); (T.H.); (Y.L.); (S.L.)
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| | - Tianzhuang Huang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China; (S.L.); (L.S.); (T.H.); (Y.L.); (S.L.)
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| | - Yuyi Luo
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China; (S.L.); (L.S.); (T.H.); (Y.L.); (S.L.)
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| | - Yongchang Chen
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China; (S.L.); (L.S.); (T.H.); (Y.L.); (S.L.)
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| | - Shangang Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China; (S.L.); (L.S.); (T.H.); (Y.L.); (S.L.)
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| | - Zhengbo Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China; (S.L.); (L.S.); (T.H.); (Y.L.); (S.L.)
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| |
Collapse
|
6
|
Liu DY, Wu Y, Feng ZQ, Yu Y, Cai HW, Liao SP, Zeng T, Zhu L, Wang X, Wan LH. Rosmarinic acid against cognitive impairment via RACK1/HIF-1α regulated microglial polarization in sepsis-surviving mice. Chem Biol Interact 2024; 388:110830. [PMID: 38103880 DOI: 10.1016/j.cbi.2023.110830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 11/29/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
Microglial polarization modulation has been considered the potential therapeutic strategy for relieving cognitive impairment in sepsis survivors. Rosmarinic acid (RA), a water-soluble polyphenolic natural compound, processes a strong protective effect on various types of neurological disorders including Parkinson's disease, depression, and anxiety. However, its role and potential molecular mechanisms in sepsis-associated cognitive impairment remain unclear. To investigate the preventive and therapeutic effect of RA on sepsis-associated cognitive impairment and elucidate the potential mechanism of RA on regulating microglial polarization, we established a CLP-induced cognitive impairment model in mice and a lipopolysaccharide-induced microglia polarization cell model in BV-2. RACK1 siRNA was designed to identify the potential molecular mechanism of RACK1 on microglial polarization. The preventive and therapeutic effect of RA on cognitive impairment followed by PET-CT and behavioral tests including open-field test and tail suspension test. RACK1/HIF-1α pathway and microglial morphology in the hippocampus or BV-2 cells were measured. The results showed that RA significantly ameliorated the CLP-induced depressive and anxiety-like behaviors and promoted whole-brain glucose uptake in mice. Moreover, RA markedly improved CLP-induced hippocampal neuron loss and microglial activation by inhibiting microglial M1 polarization. Furthermore, experiments showed RACK1 was involved in the regulation of LPS-induced microglial M1 polarization via HIF-1α, and RA suppressed lipopolysaccharide or sepsis-associated microglial M1 polarization via RACK1/HIF-1α pathway (rescued the decrease of RACK1 and increase of HIF-1α). Taken together, RA could be a potential preventive and therapeutic medication in improving cognitive impairment through RACK1/HIF-1α pathway-regulated microglial polarization.
Collapse
Affiliation(s)
- Dan-Yang Liu
- NHC Key Laboratory of Chronobiology (Sichuan University), West China School of Basic Medical Sciences & Forensic Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China; Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Yao Wu
- NHC Key Laboratory of Chronobiology (Sichuan University), West China School of Basic Medical Sciences & Forensic Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China; Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Zi-Qi Feng
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, PR China; Top-notch Undergraduate Training Program 2.0, Grade 2019, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, PR China
| | - Yang Yu
- NHC Key Laboratory of Chronobiology (Sichuan University), West China School of Basic Medical Sciences & Forensic Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China; Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Hua-Wei Cai
- Department of Nuclear Medicine and Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Shi-Ping Liao
- Functional Laboratory, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Tao Zeng
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Ling Zhu
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Xia Wang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, 610041, PR China.
| | - Li-Hong Wan
- NHC Key Laboratory of Chronobiology (Sichuan University), West China School of Basic Medical Sciences & Forensic Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China; Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, PR China.
| |
Collapse
|
7
|
Pappolla MA, Wu P, Fang X, Poeggeler B, Sambamurti K, Wisniewski T, Perry G. Stem Cell Interventions in Neurology: From Bench to Bedside. J Alzheimers Dis 2024; 101:S395-S416. [PMID: 39422938 DOI: 10.3233/jad-230897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Stem cell therapies are progressively redefining the treatment landscape for a spectrum of neurological and age-related disorders. This review discusses the molecular and functional attributes of stem cells, emphasizing the roles of neural stem cells and mesenchymal stem cells in the context of neurological diseases such as stroke, multiple sclerosis, amyotrophic lateral sclerosis, traumatic brain injury, Parkinson's disease, and Alzheimer's disease. The review also explores the potential of stem cells in addressing the aging process. The paper analyzes stem cells' intrinsic properties of self-renewal, differentiation, and paracrine effects, alongside the importance of laboratory-modified stem cells like induced pluripotent stem cells and transgenic stem cells. Insights into disease-specific stem cell treatments are offered, reviewing both successes and challenges in the field. This includes the translational difficulties from rodent studies to human trials. The review concludes by acknowledging the uncharted territories that warrant further investigation, emphasizing the potential roles of stem cell-derived exosomes and indole-related molecules, and aiming at providing a basic understanding of stem cell therapies.
Collapse
Affiliation(s)
- Miguel A Pappolla
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Ping Wu
- Department of Neuroscience & Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Xiang Fang
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Burkhard Poeggeler
- Johann-Friedrich-Blumenbach-Institute for Zoology and Anthropology, Faculty of Biology and Psychology, Georg August University Göttingen, Gütersloh, Germany
| | - Kumar Sambamurti
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA
| | - Thomas Wisniewski
- Departments of Neurology, Pathology, and Psychiatry, New York University Alzheimer's Research Center, New York University Grossman School of Medicine, New York, NY, USA
| | - George Perry
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas at San Antonio, San Antonio, TX, USA
| |
Collapse
|
8
|
Sanker V, Kundu M, El Kassem S, El Nouiri A, Emara M, Maaz ZA, Nazir A, Bekele BK, Uwishema O. Posttraumatic hydrocephalus: Recent advances and new therapeutic strategies. Health Sci Rep 2023; 6:e1713. [PMID: 38028696 PMCID: PMC10652704 DOI: 10.1002/hsr2.1713] [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: 07/02/2023] [Revised: 10/28/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023] Open
Abstract
Background Hydrocephalus or ventriculomegaly is a condition brought on by an overabundance of cerebrospinal fluid (CSF) in the ventricular system. The major contributor to posttraumatic hydrocephalus (PTH) is traumatic brain injuries (TBIs), especially in individuals with occupations set in industrial settings. A variety of criteria have been employed for the diagnosis of PTH, including the combination of neurological symptoms like nerve deficits and headache, as well as an initial improvement followed by a worsened relapse of altered consciousness and neurological deterioration, which is detected by computed tomography-brain imaging that reveals gradual ventriculomegaly. Aim In this article, we discuss and summarize briefly the current understandings and advancements in the management of PTH. Methods The available literature for this review was searched on various bibliographic databases using an individually verified, prespecified approach. The level of evidence of the included studies was considered as per the Centre for Evidence-Based Medicine recommendations. Results The commonly practiced current treatment modality involves shunting CSF but is often associated with complications and recurrence. The lack of a definitive management strategy for PTH warrants the utilization of novel and innovative modalities such as stem cell transplantations and antioxidative stress therapies. Conclusion One of the worst complications of a TBI is PTH, which has a high morbidity and mortality rate. Even though there hasn't been a successful method in stopping PTH from happening, hemorrhage-derived blood, and its metabolic by-products, like iron, hemoglobin, free radicals, thrombin, and red blood cells, may be potential targets for PTH hindrance and management. Also, using stem cell transplantations in animal models and antioxidative stress therapies in future studies can lower PTH occurrence and improve its outcome. Moreover, the integration of clinical trials and theoretical knowledge should be encouraged in future research projects to establish effective and updated management guidelines for PTH.
Collapse
Affiliation(s)
- Vivek Sanker
- Oli Health Magazine Organization, Research and EducationKigaliRwanda
- Society of Brain Mapping and TherapeuticsLos AngelesCaliforniaUSA
| | - Mrinmoy Kundu
- Oli Health Magazine Organization, Research and EducationKigaliRwanda
- Institute of Medical Sciences and SUM HospitalBhubaneswarIndia
| | - Sarah El Kassem
- Oli Health Magazine Organization, Research and EducationKigaliRwanda
- Faculty of MedicineBeirut Arab UniversityBeirutLebanon
| | - Ahmad El Nouiri
- Oli Health Magazine Organization, Research and EducationKigaliRwanda
- Faculty of MedicineBeirut Arab UniversityBeirutLebanon
| | - Mohamed Emara
- Oli Health Magazine Organization, Research and EducationKigaliRwanda
- College of MedicineUniversity of SharjahSharjahUnited Arab Emirates
| | - Zeina Al Maaz
- Oli Health Magazine Organization, Research and EducationKigaliRwanda
- Faculty of MedicineBeirut Arab UniversityBeirutLebanon
| | - Abubakar Nazir
- Oli Health Magazine Organization, Research and EducationKigaliRwanda
| | - Bezawit Kassahun Bekele
- Oli Health Magazine Organization, Research and EducationKigaliRwanda
- School of MedicineAddis Ababa UniversityAddis AbabaEthiopia
- Milken Institute of Public HealthGeorge Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Olivier Uwishema
- Oli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of medicineClinton Global Initiative UniversityNew YorkNew YorkUSA
- Faculty of MedicineKaradeniz Technical UniversityTrabzonTurkey
| |
Collapse
|
9
|
Schellino R, Besusso D, Parolisi R, Gómez-González GB, Dallere S, Scaramuzza L, Ribodino M, Campus I, Conforti P, Parmar M, Boido M, Cattaneo E, Buffo A. hESC-derived striatal progenitors grafted into a Huntington's disease rat model support long-term functional motor recovery by differentiating, self-organizing and connecting into the lesioned striatum. Stem Cell Res Ther 2023; 14:189. [PMID: 37507794 PMCID: PMC10386300 DOI: 10.1186/s13287-023-03422-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Huntington's disease (HD) is a motor and cognitive neurodegenerative disorder due to prominent loss of striatal medium spiny neurons (MSNs). Cell replacement using human embryonic stem cells (hESCs) derivatives may offer new therapeutic opportunities to replace degenerated neurons and repair damaged circuits. METHODS With the aim to develop effective cell replacement for HD, we assessed the long-term therapeutic value of hESC-derived striatal progenitors by grafting the cells into the striatum of a preclinical model of HD [i.e., adult immunodeficient rats in which the striatum was lesioned by monolateral injection of quinolinic acid (QA)]. We examined the survival, maturation, self-organization and integration of the graft as well as its impact on lesion-dependent motor alterations up to 6 months post-graft. Moreover, we tested whether exposing a cohort of QA-lesioned animals to environmental enrichment (EE) could improve graft integration and function. RESULTS Human striatal progenitors survived up to 6 months after transplantation and showed morphological and neurochemical features typical of human MSNs. Donor-derived interneurons were also detected. Grafts wired in both local and long-range striatal circuits, formed domains suggestive of distinct ganglionic eminence territories and displayed emerging striosome features. Moreover, over time grafts improved complex motor performances affected by QA. EE selectively increased cell differentiation into MSN phenotype and promoted host-to-graft connectivity. However, when combined to the graft, the EE paradigm used in this study was insufficient to produce an additive effect on task execution. CONCLUSIONS The data support the long-term therapeutic potential of ESC-derived human striatal progenitor grafts for the replacement of degenerated striatal neurons in HD and suggest that EE can effectively accelerate the maturation and promote the integration of human striatal cells.
Collapse
Affiliation(s)
- Roberta Schellino
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126, Turin, Italy.
- Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043, Orbassano, Italy.
| | - Dario Besusso
- Department of Biosciences, University of Milan, 20122, Milan, Italy
- National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", 20133, Milan, Italy
| | - Roberta Parolisi
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043, Orbassano, Italy
| | - Gabriela B Gómez-González
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043, Orbassano, Italy
| | - Sveva Dallere
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043, Orbassano, Italy
| | - Linda Scaramuzza
- Department of Biosciences, University of Milan, 20122, Milan, Italy
- National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", 20133, Milan, Italy
| | - Marta Ribodino
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043, Orbassano, Italy
| | - Ilaria Campus
- Department of Biosciences, University of Milan, 20122, Milan, Italy
- National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", 20133, Milan, Italy
| | - Paola Conforti
- Department of Biosciences, University of Milan, 20122, Milan, Italy
- National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", 20133, Milan, Italy
| | - Malin Parmar
- Wallenberg Neuroscience Center and Lund Stem Cell Center, Lund University, 22184, Lund, Sweden
| | - Marina Boido
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043, Orbassano, Italy
| | - Elena Cattaneo
- Department of Biosciences, University of Milan, 20122, Milan, Italy
- National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", 20133, Milan, Italy
| | - Annalisa Buffo
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126, Turin, Italy.
- Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043, Orbassano, Italy.
| |
Collapse
|
10
|
Chen T, Xia Y, Zhang L, Xu T, Yi Y, Chen J, Liu Z, Yang L, Chen S, Zhou X, Chen X, Wu H, Liu J. Loading neural stem cells on hydrogel scaffold improves cell retention rate and promotes functional recovery in traumatic brain injury. Mater Today Bio 2023; 19:100606. [PMID: 37063247 PMCID: PMC10102240 DOI: 10.1016/j.mtbio.2023.100606] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/23/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Neural stem cell (NSC) has gained considerable attention in traumatic brain injury (TBI) treatment because of their ability to replenish dysfunctional neurons and stimulate endogenous neurorestorative processes. However, their therapeutic effects are hindered by the low cell retention rate after transplantation into the dynamic brain. In this study, we found cerebrospinal fluid (CSF) flow after TBI is an important factor associated with cell loss following NSC transplantation. Recently, several studies have shown that hydrogels could serve as a beneficial carrier for stem cell transplantation, which provides a solution to prevent CSF flow-induced cell loss after TBI. For this purpose, we evaluated three different hydrogel scaffolds and found the gelatin methacrylate (GelMA)/sodium alginate (Alg) (GelMA/Alg) hydrogel scaffold showed the best capabilities for NSC adherence, growth, and differentiation. Additionally, we detected that pre-differentiated NSCs, which were loaded on the GelMA/Alg hydrogel and cultured for 7 days in neuronal differentiation medium (NSC [7d]), had the highest cell retention rate after CSF impact. Next, the neuroprotective effects of the NSC-loaded GelMA/Alg hydrogel scaffold were evaluated in a rat model of TBI. NSC [7d]-loaded GelMA/Alg markedly decreased microglial activation and neuronal death in the acute phase, reduced tissue loss, alleviated astrogliosis, promoted neurogenesis, and improved neurological recovery in the chronic phase. In summary, we demonstrated that the integration with the GelMA/Alg and modification of NSC differentiation could inhibit the influence of CSF flow on transplanted NSCs, leading to increased number of retained NSCs and improved neuroprotective effects, providing a promising alternative for TBI treatment.
Collapse
Affiliation(s)
- Tiange Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuguo Xia
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tao Xu
- Bio-Intelligent Manufacturing and Living Matter Bioprinting Center, Research Institute of Tsinghua University in Shenzhen, Tsinghua University, Shenzhen, China
| | - Yan Yi
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Hunan, China
| | - Jianwei Chen
- Bio-Intelligent Manufacturing and Living Matter Bioprinting Center, Research Institute of Tsinghua University in Shenzhen, Tsinghua University, Shenzhen, China
| | - Ziyuan Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liting Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Siming Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoxi Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haiyu Wu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jinfang Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Corresponding author. Department of Neurosurgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Rd, Kaifu District, Changsha, 410008, PR China.
| |
Collapse
|
11
|
de Almeida MMA, Goodkey K, Voronova A. Regulation of microglia function by neural stem cells. Front Cell Neurosci 2023; 17:1130205. [PMID: 36937181 PMCID: PMC10014810 DOI: 10.3389/fncel.2023.1130205] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/13/2023] [Indexed: 03/05/2023] Open
Abstract
Neural stem and precursor cells (NPCs) build and regenerate the central nervous system (CNS) by maintaining their pool (self-renewal) and differentiating into neurons, astrocytes, and oligodendrocytes (multipotency) throughout life. This has inspired research into pro-regenerative therapies that utilize transplantation of exogenous NPCs or recruitment of endogenous adult NPCs for CNS regeneration and repair. Recent advances in single-cell RNA sequencing and other "omics" have revealed that NPCs express not just traditional progenitor-related genes, but also genes involved in immune function. Here, we review how NPCs exert immunomodulatory function by regulating the biology of microglia, immune cells that are present in NPC niches and throughout the CNS. We discuss the role of transplanted and endogenous NPCs in regulating microglia fates, such as survival, proliferation, migration, phagocytosis and activation, in the developing, injured and degenerating CNS. We also provide a literature review on NPC-specific mediators that are responsible for modulating microglia biology. Our review highlights the immunomodulatory properties of NPCs and the significance of these findings in the context of designing pro-regenerative therapies for degenerating and diseased CNS.
Collapse
Affiliation(s)
- Monique M. A. de Almeida
- Department of Medical Genetics, Faculty of Medicine & Dentistry, Edmonton, AB, Canada
- Faculty of Medicine & Dentistry, Neuroscience and Mental Health Institute, Edmonton, AB, Canada
| | - Kara Goodkey
- Department of Medical Genetics, Faculty of Medicine & Dentistry, Edmonton, AB, Canada
- Women and Children’s Health Research Institute, 5-083 Edmonton Clinic Health Academy, University of Alberta, Edmonton, AB, Canada
| | - Anastassia Voronova
- Department of Medical Genetics, Faculty of Medicine & Dentistry, Edmonton, AB, Canada
- Faculty of Medicine & Dentistry, Neuroscience and Mental Health Institute, Edmonton, AB, Canada
- Women and Children’s Health Research Institute, 5-083 Edmonton Clinic Health Academy, University of Alberta, Edmonton, AB, Canada
- Department of Cell Biology, Faculty of Medicine & Dentistry, Edmonton, AB, Canada
- Multiple Sclerosis Centre and Department of Cell Biology, Faculty of Medicine & Dentistry, Edmonton, AB, Canada
| |
Collapse
|
12
|
Fan H, Duan H, Hao P, Gao Y, Zhao W, Hao F, Li X, Yang Z. Cellular regeneration treatments for traumatic brain injury. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
13
|
Stem Cell Therapy for Sequestration of Traumatic Brain Injury-Induced Inflammation. Int J Mol Sci 2022; 23:ijms231810286. [PMID: 36142198 PMCID: PMC9499317 DOI: 10.3390/ijms231810286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/30/2022] [Accepted: 09/04/2022] [Indexed: 11/17/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of long-term neurological disabilities in the world. TBI is a signature disease for soldiers and veterans, but also affects civilians, including adults and children. Following TBI, the brain resident and immune cells turn into a “reactive” state, characterized by the production of inflammatory mediators that contribute to the development of cognitive deficits. Other injuries to the brain, including radiation exposure, may trigger TBI-like pathology, characterized by inflammation. Currently there are no treatments to prevent or reverse the deleterious consequences of brain trauma. The recognition that TBI predisposes stem cell alterations suggests that stem cell-based therapies stand as a potential treatment for TBI. Here, we discuss the inflamed brain after TBI and radiation injury. We further review the status of stem cells in the inflamed brain and the applications of cell therapy in sequestering inflammation in TBI.
Collapse
|
14
|
Qin Q, Wang T, Xu Z, Liu S, Zhang H, Du Z, Wang J, Wang Y, Wang Z, Yuan S, Wu J, He W, Wang C, Yan X, Wang Y, Jiang X. Ectoderm-derived frontal bone mesenchymal stem cells promote traumatic brain injury recovery by alleviating neuroinflammation and glutamate excitotoxicity partially via FGF1. Stem Cell Res Ther 2022; 13:341. [PMID: 35883153 PMCID: PMC9327213 DOI: 10.1186/s13287-022-03032-6] [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: 12/22/2021] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Background Traumatic brain injury (TBI) leads to cell and tissue impairment, as well as functional deficits. Stem cells promote structural and functional recovery and thus are considered as a promising therapy for various nerve injuries. Here, we aimed to investigate the role of ectoderm-derived frontal bone mesenchymal stem cells (FbMSCs) in promoting cerebral repair and functional recovery in a murine TBI model. Methods A murine TBI model was established by injuring C57BL/6 N mice with moderate-controlled cortical impact to evaluate the extent of brain damage and behavioral deficits. Ectoderm-derived FbMSCs were isolated from the frontal bone and their characteristics were assessed using multiple differentiation assays, flow cytometry and microarray analysis. Brain repairment and functional recovery were analyzed at different days post-injury with or without FbMSC application. Behavioral tests were performed to assess learning and memory improvements. RNA sequencing analysis, immunofluorescence staining, and quantitative reverse-transcription polymerase chain reaction (qRT-PCR) were used to examine inflammation reaction and neural regeneration. In vitro co-culture analysis and quantification of glutamate transportation were carried out to explore the possible mechanism of neurogenesis and functional recovery promoted by FbMSCs. Results Ectoderm-derived FbMSCs showed fibroblast like morphology and osteogenic differentiation capacity. FbMSCs were CD105, CD29 positive and CD45, CD31 negative. Different from mesoderm-derived MSCs, FbMSCs expressed the ectoderm-specific transcription factor Tfap2β. TBI mice showed impaired learning and memory deficits. Microglia and astrocyte activation, as well as neural damage, were significantly increased post-injury. FbMSC application ameliorated the behavioral deficits of TBI mice and promoted neural regeneration. RNA sequencing analysis showed that signal pathways related to inflammation decreased, whereas those related to neural activation increased. Immunofluorescence staining and qRT-PCR data revealed that microglial activation and astrocyte polarization to the A1 phenotype were suppressed by FbMSC application. In addition, FGF1 secreted from FbMSCs enhanced glutamate transportation by astrocytes and alleviated the cytotoxic effect of excessive glutamate on neurons. Conclusions Ectoderm-derived FbMSC application significantly alleviated neuroinflammation, brain injury, and excitatory toxicity to neurons, improved cognition and behavioral deficits in TBI mice. Therefore, ectoderm-derived FbMSCs could be ideal therapeutic candidates for TBI which mostly affect cells from the same embryonic origins as FbMSCs. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03032-6.
Collapse
Affiliation(s)
- Qiaozhen Qin
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China.,Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, People's Republic of China
| | - Ting Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Zhenhua Xu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Shuirong Liu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Heyang Zhang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Zhangzhen Du
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Jianing Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Yadi Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Zhenning Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Shanshan Yuan
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Jiamei Wu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Wenyan He
- China National Clinical Research Center for Neurological Diseases, Jing-Jin Center for Neuroinflammation, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Changzhen Wang
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Xinlong Yan
- Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, People's Republic of China.
| | - Yan Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China. .,Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China.
| | - Xiaoxia Jiang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China. .,Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China.
| |
Collapse
|
15
|
Modulating Microglia/Macrophage Activation by CDNF Promotes Transplantation of Fetal Ventral Mesencephalic Graft Survival and Function in a Hemiparkinsonian Rat Model. Biomedicines 2022; 10:biomedicines10061446. [PMID: 35740467 PMCID: PMC9221078 DOI: 10.3390/biomedicines10061446] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/08/2022] [Accepted: 06/11/2022] [Indexed: 11/30/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by the loss of dopaminergic neurons in substantia nigra pars compacta, which leads to the motor control deficits. Recently, cell transplantation is a cutting-edge technique for the therapy of PD. Nevertheless, one key bottleneck to realizing such potential is allogenic immune reaction of tissue grafts by recipients. Cerebral dopamine neurotrophic factor (CDNF) was shown to possess immune-modulatory properties that benefit neurodegenerative diseases. We hypothesized that co-administration of CDNF with fetal ventral mesencephalic (VM) tissue can improve the success of VM replacement therapies by attenuating immune responses. Hemiparkinsonian rats were generated by injecting 6-hydroxydopamine (6-OHDA) into the right medial forebrain bundle of Sprague Dawley (SD) rats. The rats were then intrastriatally transplanted with VM tissue from rats, with/without CDNF administration. Recovery of dopaminergic function and survival of the grafts were evaluated using the apomorphine-induced rotation test and small-animal positron emission tomography (PET) coupled with [18F] DOPA or [18F] FE-PE2I, respectively. In addition, transplantation-related inflammatory response was determined by uptake of [18F] FEPPA in the grafted side of striatum. Immunohistochemistry (IHC) examination was used to determine the survival of the grated dopaminergic neurons in the striatum and to investigate immune-modulatory effects of CDNF. The modulation of inflammatory responses caused by CDNF might involve enhancing M2 subset polarization and increasing fractal dimensions of 6-OHDA-treated BV2 microglial cell line. Analysis of CDNF-induced changes to gene expressions of 6-OHDA-stimulated BV2 cells implies that these alternations of the biomarkers and microglial morphology are implicated in the upregulation of protein kinase B signaling as well as regulation of catalytic, transferase, and protein serine/threonine kinase activity. The effects of CDNF on 6-OHDA-induced alternation of the canonical pathway in BV2 microglial cells is highly associated with PI3K-mediated phagosome formation. Our results are the first to show that CDNF administration enhances the survival of the grafted dopaminergic neurons and improves functional recovery in PD animal model. Modulation of the polarization, morphological characteristics, and transcriptional profiles of 6-OHDA-stimualted microglia by CDNF may possess these properties in transplantation-based regenerative therapies.
Collapse
|
16
|
Xiong Y, Song J, Huang X, Pan Z, Goldbrunner R, Stavrinou L, Lin S, Hu W, Zheng F, Stavrinou P. Exosomes Derived From Mesenchymal Stem Cells: Novel Effects in the Treatment of Ischemic Stroke. Front Neurosci 2022; 16:899887. [PMID: 35585925 PMCID: PMC9108502 DOI: 10.3389/fnins.2022.899887] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/13/2022] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke is defined as an infarction in the brain, caused by impaired cerebral blood supply, leading to local brain tissue ischemia, hypoxic necrosis, and corresponding neurological deficits. At present, revascularization strategies in patients with acute ischemic stroke include intravenous thrombolysis and mechanical endovascular treatment. However, due to the short treatment time window (<4.5 h) and method restrictions, clinical research is focused on new methods to treat ischemic stroke. Exosomes are nano-sized biovesicles produced in the endosomal compartment of most eukaryotic cells, containing DNA, complex RNA, and protein (30-150 nm). They are released into surrounding extracellular fluid upon fusion between multivesicular bodies and the plasma membrane. Exosomes have the characteristics of low immunogenicity, good innate stability, high transmission efficiency, and the ability to cross the blood-brain barrier, making them potential therapeutic modalities for the treatment of ischemic stroke. The seed sequence of miRNA secreted by exosomes is base-paired with complementary mRNA to improve the microenvironment of ischemic tissue, thereby regulating downstream signal transduction activities. With exosome research still in the theoretical and experimental stages, this review aims to shed light on the potential of exosomes derived from mesenchymal stem cells in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yu Xiong
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Jianping Song
- Department of Neurosurgery, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
- Department of Neurosurgery, National Regional Medical Center, Fudan University Huashan Hospital Fujian Campus, The First Affiliated Hospital Binhai Campus, Fujian Medical University, Fuzhou, China
| | - Xinyue Huang
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Zhigang Pan
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Roland Goldbrunner
- Department of Neurosurgery, Faculty of Medicine and University Hospital, Center for Neurosurgery, University of Cologne, Cologne, Germany
| | - Lampis Stavrinou
- 2nd Department of Neurosurgery, Athens Medical School, “Attikon” University Hospital, National and Kapodistrian University, Athens, Greece
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Weipeng Hu
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Feng Zheng
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Pantelis Stavrinou
- Department of Neurosurgery, Faculty of Medicine and University Hospital, Center for Neurosurgery, University of Cologne, Cologne, Germany
- Department of Neurosurgery, Metropolitan Hospital, Athens, Greece
| |
Collapse
|
17
|
Pedachenko EG, Liubich LD, Staino LP, Egorova DM, Skaterna TD. Neuroregenerative “Bystander”-Effects of Conditioned Media from Adipose Tissue-Derived Fibroblast-Like Cells in Vitro. CYTOL GENET+ 2022. [DOI: 10.3103/s0095452722020098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
18
|
Neural stem cell therapy in conjunction with curcumin loaded in niosomal nanoparticles enhanced recovery from traumatic brain injury. Sci Rep 2022; 12:3572. [PMID: 35246564 PMCID: PMC8897489 DOI: 10.1038/s41598-022-07367-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 02/16/2022] [Indexed: 12/28/2022] Open
Abstract
Despite a great amount of effort, there is still a need for reliable treatments of traumatic brain injury (TBI). Recently, stem cell therapy has emerged as a new avenue to address neuronal regeneration after TBI. However, the environment of TBI lesions exerts negative effects on the stem cells efficacy. Therefore, to maximize the beneficial effects of stem cells in the course of TBI, we evaluated the effect of human neural stem/progenitor cells (hNS/PCs) and curcumin-loaded niosome nanoparticles (CM-NPs) on behavioral changes, brain edema, gliosis, and inflammatory responses in a rat model of TBI. After TBI, hNS/PCs were transplanted within the injury site and CM-NPs were orally administered for 10 days. Finally, the effect of combination therapy was compared to several control groups. Our results indicated a significant improvement of general locomotor activity in the hNS/PCs + CM-NPs treatment group compared to the control groups. We also observed a significant improvement in brain edema in the hNS/PCs + CM-NPs treatment group compared to the other groups. Furthermore, a significant decrease in astrogliosis was seen in the combined treatment group. Moreover, TLR4-, NF-κB-, and TNF-α- positive cells were significantly decreased in hNS/PCs + CM-NPs group compared to the control groups. Taken together, this study indicated that combination therapy of stem cells with CM-NPs can be an effective therapy for TBI.
Collapse
|
19
|
Xu L, Wang J, Ding Y, Wang L, Zhu YJ. Current Knowledge of Microglia in Traumatic Spinal Cord Injury. Front Neurol 2022; 12:796704. [PMID: 35087472 PMCID: PMC8787368 DOI: 10.3389/fneur.2021.796704] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/07/2021] [Indexed: 12/12/2022] Open
Abstract
Microglia are the resident immune cells in the central nervous system (CNS). After traumatic spinal cord injury (SCI), microglia undergo activation, proliferation, and changes in gene and protein expression and morphology, with detrimental and beneficial effects. Activated microglia cause secondary neuronal injury via the production of proinflammatory cytokines, reactive oxygen species, and proteases. However, activated microglia also promote neuronal repair through the secretion of anti-inflammatory growth factors and cytokines. Proinflammatory cytokines increase endothelial permeability, promote A1 astrocyte activation and axonal demyelination, and reduce neural stem/progenitor cells (NSPCs), leading to the exacerbation of neuronal injury. In contrast, anti-inflammatory factors facilitate angiogenesis, reduce reactive astrocytes, and promote axonal remyelination and the propagation of NSPCs, contributing to tissue repair and locomotor recovery. Due to its limited regenerative capacity, the CNS requires beneficial microglia for continuous protection against injury. Understanding and regulating microglial activation status are beneficial to reducing detrimental effects and promoting repair behaviors and to obtain more information on efficient therapies for traumatic SCI. This review discusses microglial activation and the differences between microglia and similar immune cells, microglial interactions with other cells in the spinal cord, and the progress in the development of therapies targeting microglia in SCI.
Collapse
Affiliation(s)
- Lintao Xu
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jingyu Wang
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yueming Ding
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Linlin Wang
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yong-Jian Zhu
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
20
|
Campos HC, Ribeiro DE, Hashiguchi D, Hukuda DY, Gimenes C, Romariz SAA, Ye Q, Tang Y, Ulrich H, Longo BM. Distinct Effects of the Hippocampal Transplantation of Neural and Mesenchymal Stem Cells in a Transgenic Model of Alzheimer's Disease. Stem Cell Rev Rep 2022; 18:781-791. [PMID: 34997526 DOI: 10.1007/s12015-021-10321-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is a severe disabling condition with no cure currently available, which accounts for 60-70% of all dementia cases worldwide. Therefore, the investigation of possible therapeutic strategies for AD is necessary. To this end, animal models corresponding to the main aspects of AD in humans have been widely used. Similar to AD patients, the double transgenic APPswe/PS1dE9 (APP/PS1) mice show cognitive deficits, hyperlocomotion, amyloid-β (Αβ) plaques in the cortex and hippocampus, and exacerbated inflammatory responses. Recent studies have shown that these neuropathological features could be reversed by stem cell transplantation. However, the effects induced by neural (NSC) and mesenchymal (MSC) stem cells has never been compared in an AD animal model. Therefore, the present study aimed to investigate whether transplantation of NSC or MSC into the hippocampus of APP/PS1 mice reverses AD-induced pathological alterations, evaluated by the locomotor activity (open field test), short- and long-term memory (object recognition) tests, Αβ plaques (6-E10), microglia distribution (Iba-1), M1 (iNOS) and M2 (ARG-1) microglial phenotype frequencies. NSC and MSC engraftment reduced the number of Αβ plaques and produced an increase in M2 microglia polarization in the hippocampus of APP/PS1 mice, suggesting an anti-inflammatory effect of stem cell transplantation. NSC also reversed the hyperlocomotor activity and increased the number of microglia in the hippocampus of APP/PS1 mice. No impairment of short or long-term memory was observed in APP/PS1 mice. Overall, this study highlights the potential beneficial effects of transplanting NSC or MSC for AD treatment.
Collapse
Affiliation(s)
- Henrique C Campos
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Deidiane Elisa Ribeiro
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil
| | - Debora Hashiguchi
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil.,Laboratório de Plasticidade Sináptica, Brain Institute, Federal University of Rio Grande do Norte, Natal, RN, 59078-900, Caixa Postal: 1524, Brazil
| | - Deborah Y Hukuda
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Christiane Gimenes
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Simone A A Romariz
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Qing Ye
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil.,International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.,Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, 610075, China
| | - Yong Tang
- International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.,Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, 610075, China
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil.,International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Beatriz Monteiro Longo
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
21
|
Wang G, Wu HL, Liu YP, Yan DQ, Yuan ZL, Chen L, Yang Q, Gao YS, Diao B. Pre-clinical study of human umbilical cord mesenchymal stem cell transplantation for the treatment of traumatic brain injury: safety evaluation from immunogenic and oncogenic perspectives. Neural Regen Res 2022; 17:354-361. [PMID: 34269210 PMCID: PMC8463980 DOI: 10.4103/1673-5374.317985] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Stem cell therapy is a promising strategy for the treatment of traumatic brain injury (TBI). However, animal experiments are needed to evaluate safety; in particular, to examine the immunogenicity and tumorigenicity of human umbilical cord mesenchymal stem cells (huMSCs) before clinical application. In this study, huMSCs were harvested from human amniotic membrane and umbilical cord vascular tissue. A rat model of TBI was established using the controlled cortical impact method. Starting from the third day after injury, the rats were injected with 10 μL of 5 × 106/mL huMSCs by cerebral stereotaxis or with 500 μL of 1 × 106/mL huMSCs via the tail vein for 3 successive days. huMSC transplantation decreased the serum levels of proinflammatory cytokines in rats with TBI and increased the serum levels of anti-inflammatory cytokines, thereby exhibiting good immunoregulatory function. The transplanted huMSCs were distributed in the liver, lung and brain injury sites. No abnormal proliferation or tumorigenesis was found in these organs up to 12 months after transplantation. The transplanted huMSCs negligibly proliferated in vivo, and apoptosis was gradually observed at later stages. These findings suggest that huMSC transplantation for the treatment of traumatic brain injury displays good safety. In addition, huMSCs exhibit good immunoregulatory function, which can help prevent and reduce secondary brain injury caused by the rapid release of inflammatory factors after TBI. This study was approved by the Ethics Committee of Wuhan General Hospital of PLA (approval No. 20160054) on November 1, 2016.
Collapse
Affiliation(s)
- Gang Wang
- Basic Medical Laboratory, General Hospital of the Central Theater Command; Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei Province, China
| | - Hua-Ling Wu
- Department of Clinical Laboratory, The Third People's Hospital of Yongzhou, Yongzhou, Hunan Province, China
| | - Yue-Ping Liu
- Basic Medical Laboratory, General Hospital of the Central Theater Command; Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei Province, China
| | - De-Qi Yan
- Department of Neurosurgery, 990th Hospital of Joint Logistic Support Troops of PLA, Zhumadian, Henan Province, China
| | - Zi-Lin Yuan
- Basic Medical Laboratory, General Hospital of the Central Theater Command; Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei Province, China
| | - Li Chen
- Basic Medical Laboratory, General Hospital of the Central Theater Command; Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei Province, China
| | - Qian Yang
- Basic Medical Laboratory, General Hospital of the Central Theater Command; Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei Province, China
| | - Yu-Song Gao
- Department of Neurosurgery, 990th Hospital of Joint Logistic Support Troops of PLA, Zhumadian, Henan Province, China
| | - Bo Diao
- Basic Medical Laboratory, General Hospital of the Central Theater Command; Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei Province, China
| |
Collapse
|
22
|
Abstract
Traumatic injury of the central nervous system (CNS) is a worldwide health problem affecting millions of people. Trauma of the CNS, that is, traumatic brain injury (TBI) and spinal cord injury (SCI), lead to massive and progressive cell loss and axonal degeneration, usually with very limited regeneration. At present, there are no treatments to protect injured CNS tissue or to replace the lost tissue. Stem cells are a cell type that by definition can self-renew and give rise to multiple cell lineages. In recent years, therapies using stem and progenitor cells have shown promising effects in experimental CNS trauma, particularly in the acute-subacute stage, but also in chronic injuries. However, the therapeutic mechanisms by which transplanted cells achieve the structural and/or functional improvements are often not clear. Stem cell therapies for CNS trauma can be categorized into 2 main concepts, transplantation of exogenous neural stem cells and neural progenitor cells and recruitment of endogenous stem and progenitor cells. In this review, focusing on the advances during the last decade, we will discuss the major cell therapies, the pros and cons of these 2 concepts for TBI and SCI, and the treatment strategies we believe will be successful.
Collapse
Affiliation(s)
- Xiaofei Li
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Erik Sundström
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Corresponding author: Erik Sundström, Department of Neurobiology, Care Sciences and Society (NVS), BioClinicum J9:20, Karolinska University Hospital, S17164 Solna, Sweden.
| |
Collapse
|
23
|
Jin N, Sha W, Gao L. Shaping the Microglia in Retinal Degenerative Diseases Using Stem Cell Therapy: Practice and Prospects. Front Cell Dev Biol 2021; 9:741368. [PMID: 34966736 PMCID: PMC8710684 DOI: 10.3389/fcell.2021.741368] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022] Open
Abstract
Retinal degenerative disease (RDD) refers to a group of diseases with retinal degeneration that cause vision loss and affect people's daily lives. Various therapies have been proposed, among which stem cell therapy (SCT) holds great promise for the treatment of RDDs. Microglia are immune cells in the retina that have two activation phenotypes, namely, pro-inflammatory M1 and anti-inflammatory M2 phenotypes. These cells play an important role in the pathological progression of RDDs, especially in terms of retinal inflammation. Recent studies have extensively investigated the therapeutic potential of stem cell therapy in treating RDDs, including the immunomodulatory effects targeting microglia. In this review, we substantially summarized the characteristics of RDDs and microglia, discussed the microglial changes and phenotypic transformation of M1 microglia to M2 microglia after SCT, and proposed future directions for SCT in treating RDDs.
Collapse
Affiliation(s)
- Ni Jin
- Senior Department of Ophthalmology, The Third Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China.,Department of Endocrinology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Weiwei Sha
- Department of Endocrinology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lixiong Gao
- Senior Department of Ophthalmology, The Third Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
24
|
Cui L, Saeed Y, Li H, Yang J. Regenerative medicine and traumatic brain injury: from stem cell to cell-free therapeutic strategies. Regen Med 2021; 17:37-53. [PMID: 34905963 DOI: 10.2217/rme-2021-0069] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Traumatic brain injury (TBI) is a serious health concern, yet there is a lack of standardized treatment to combat its long-lasting effects. The objective of the present study was to provide an overview of the limitation of conventional stem-cell therapy in the treatment of TBI and to discuss the application of novel acellular therapies and their advanced strategies to enhance the efficacy of stem cells derived therapies in the light of published study data. Moreover, we also discussed the factor to optimize the therapeutic efficiency of stem cell-derived acellular therapy by overcoming the challenges for its clinical translation. Hence, we concluded that acellular therapy possesses the potential to bring a breakthrough in the field of regenerative medicine to treat TBI.
Collapse
Affiliation(s)
- Lianxu Cui
- Department of Neurosurgery, The First People's Hospital of Foshan, 81 North Lingnan Road, Foshan, Guangdong, 528300, PR China
| | - Yasmeen Saeed
- Guangdong VitaLife Biotechnology Co., LTD, 61 Xiannan Road, Nanhai District, Foshan, Guangdong, 528200, PR China
| | - Haomin Li
- Department of Neurosurgery, The First People's Hospital of Foshan, 81 North Lingnan Road, Foshan, Guangdong, 528300, PR China
| | - Jingli Yang
- School of medicine, Foshan University, 18 Jiangwan Road, Foshan, Guangdong, 528000, PR China
| |
Collapse
|
25
|
Guo Y, Meng Y, Li J, Wang H, Guo J. Effects of Bone Marrow Stromal Cells (BMSCs) on Behavior, Infarct Size and HIF-1α Expression in Stroke Rats. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study aims to assess BMSCs’ effect on the behavior, infarct size and HIF-1α expression in stroke rats. Rats were separated into sham group, CVA group and BMSCs group with 10 rats in each group followed by analysis of neuroethology scores, brain tissue pathology
and infarct size, and HIF-1α level in brain tissues. No difference of neurological scores was found between CVA group and BMSCs group after 3 hours (P > 0.05). After BMSCs transplantation, the nerve score was significantly reduced (P < 0.05) and cognitive function
was significantly improved compared to CVA group. Compared with sham rats, CAV rats had a larger area of infarction and the infarcted tissue cells showed degeneration or necrosis with reduced cell number and obvious edema, which were all improved in BMSCs group. CVA group showed a larger area
of infarct tissue (P < 0.05), which was reduced in BMSCs group (P < 0.05). Compared with sham group, CVA group showed significantly upregulated HIF-1α level (P < 0.05) which was reduced in BMSCs group (P < 0.05). BMSCs has a certain repair
effect on the ethology of stroke rats possibly via inhibition of HIF-1α level in cerebral infarction and brain tissue.
Collapse
Affiliation(s)
- Yingli Guo
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Yanbin Meng
- Department of Surgical, General Hospital of TISCO, Taiyuan, Shanxi, 030003, China
| | - Jun Li
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Hongsheng Wang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Junhong Guo
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| |
Collapse
|
26
|
Sowers JL, Sowers ML, Shavkunov AS, Hawkins BE, Wu P, DeWitt DS, Prough DS, Zhang K. Traumatic brain injury induces region-specific glutamate metabolism changes as measured by multiple mass spectrometry methods. iScience 2021; 24:103108. [PMID: 34622161 PMCID: PMC8479783 DOI: 10.1016/j.isci.2021.103108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 06/14/2021] [Accepted: 09/08/2021] [Indexed: 11/02/2022] Open
Abstract
The release of excess glutamate following traumatic brain injury (TBI) results in glutamate excitotoxicity and metabolic energy failure. Endogenous mechanisms for reducing glutamate concentration in the brain parenchyma following TBI are poorly understood. Using multiple mass spectrometry approaches, we examined TBI-induced changes to glutamate metabolism. We present evidence that glutamate concentration can be reduced by glutamate oxidation via a "truncated" tricarboxylic acid cycle coupled to the urea cycle. This process reduces glutamate levels, generates carbon for energy metabolism, leads to citrulline accumulation, and produces nitric oxide. Several key metabolites are identified by metabolomics in support of this mechanism and the locations of these metabolites in the injured hemisphere are demonstrated by MALDI-MS imaging. The results of this study establish the advantages of multiple mass spectrometry approaches and provide insights into glutamate metabolism following TBI that could lead to improved treatment approaches.
Collapse
Affiliation(s)
- James L Sowers
- MD-PhD Combined Degree Program, University of Texas Medical Branch, Galveston, TX 77555, USA.,Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX 77555, USA.,The Moody Project for Translational Traumatic Brain Injury Research, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mark L Sowers
- MD-PhD Combined Degree Program, University of Texas Medical Branch, Galveston, TX 77555, USA.,Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Alexander S Shavkunov
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Bridget E Hawkins
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555, USA.,The Moody Project for Translational Traumatic Brain Injury Research, University of Texas Medical Branch, Galveston, TX 77555, USA.,Research Innovation and Scientific Excellence (RISE) Center, School of Nursing, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ping Wu
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX 77555, USA.,The Moody Project for Translational Traumatic Brain Injury Research, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Douglas S DeWitt
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555, USA.,The Moody Project for Translational Traumatic Brain Injury Research, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Donald S Prough
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555, USA.,The Moody Project for Translational Traumatic Brain Injury Research, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Kangling Zhang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA.,The Moody Project for Translational Traumatic Brain Injury Research, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
27
|
Brousse B, Mercier O, Magalon K, Daian F, Durbec P, Cayre M. Endogenous neural stem cells modulate microglia and protect against demyelination. Stem Cell Reports 2021; 16:1792-1804. [PMID: 34087164 PMCID: PMC8282429 DOI: 10.1016/j.stemcr.2021.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 01/01/2023] Open
Abstract
In response to corpus callosum (CC) demyelination, subventricular zone-derived neural progenitors (SVZdNPs) are mobilized and generate new myelinating oligodendrocytes (OLG). Here, we examine the putative immunomodulatory properties of endogenous SVZdNPs during demyelination in the cuprizone model. SVZdNP density was higher in the lateral and rostral CC regions, and demyelination was inversely correlated with activated microglial density and pro-inflammatory cytokine levels. Single-cell RNA sequencing showed that CC areas with high levels of SVZdNP mobilization were enriched in a microglial cell subpopulation with an immunomodulatory signature. We propose MFGE8 (milk fat globule-epidermal growth factor-8) and β3 integrin as a ligand/receptor pair involved in dialogue between SVZdNPs and microglia. Immature SVZdNPs mobilized to the demyelinated CC were found highly enriched in MFGE8, which promoted the phagocytosis of myelin debris in vitro. Overall, these results demonstrate that, in addition to their cell replacement capacity, endogenous progenitors have immunomodulatory properties, highlighting a new role for endogenous SVZdNPs in myelin regeneration. Demyelination is limited in corpus callosum areas rich in subventricular zone–derived progenitors In these areas microglial cells adopt an immunomodulatory phenotype Mobilized SVZ progenitors secrete MFGE8, which promotes myelin debris phagocytosis SVZ-derived progenitors minimize demyelination by modulating microglial activity
Collapse
Affiliation(s)
- Béatrice Brousse
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), IBDM-UMR 7288, Case 907, Parc Scientifique de Luminy, Marseille Cedex 09 13288, France
| | - Océane Mercier
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), IBDM-UMR 7288, Case 907, Parc Scientifique de Luminy, Marseille Cedex 09 13288, France
| | - Karine Magalon
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), IBDM-UMR 7288, Case 907, Parc Scientifique de Luminy, Marseille Cedex 09 13288, France
| | - Fabrice Daian
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), IBDM-UMR 7288, Case 907, Parc Scientifique de Luminy, Marseille Cedex 09 13288, France
| | - Pascale Durbec
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), IBDM-UMR 7288, Case 907, Parc Scientifique de Luminy, Marseille Cedex 09 13288, France
| | - Myriam Cayre
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), IBDM-UMR 7288, Case 907, Parc Scientifique de Luminy, Marseille Cedex 09 13288, France.
| |
Collapse
|
28
|
Microglia: A Potential Drug Target for Traumatic Axonal Injury. Neural Plast 2021; 2021:5554824. [PMID: 34093701 PMCID: PMC8163545 DOI: 10.1155/2021/5554824] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/06/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
Traumatic axonal injury (TAI) is a major cause of death and disability among patients with severe traumatic brain injury (TBI); however, no effective therapies have been developed to treat this disorder. Neuroinflammation accompanying microglial activation after TBI is likely to be an important factor in TAI. In this review, we summarize the current research in this field, and recent studies suggest that microglial activation plays an important role in TAI development. We discuss several drugs and therapies that may aid TAI recovery by modulating the microglial phenotype following TBI. Based on the findings of recent studies, we conclude that the promotion of active microglia to the M2 phenotype is a potential drug target for the treatment of TAI.
Collapse
|
29
|
Jia Y, Wang G, Ye Y, Kang E, Chen H, Guo Z, He X. Niche Cells Crosstalk In Neuroinflammation After Traumatic Brain Injury. Int J Biol Sci 2021; 17:368-378. [PMID: 33390856 PMCID: PMC7757042 DOI: 10.7150/ijbs.52169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is recognized as the disease with high morbidity and disability around world in spite of the work ongoing in neural protection. Due to heterogeneity among the patients, it's still hard to acquire satisfying achievements in clinic. Neuroinflammation, which exists since primary injury occurs, with elusive duality, appear to be of significance from recovery of injury to neurogenesis. In recent years, studied have revealed that communication in neurogenic niche is more than “cell to cell” communication, and study on NSCs represent it as central role in the progress of neural regeneration. Hence, the neuroinflammation-affecting crosstalk after TBI, and clarifying definitive role of NSCs in the course of regeneration is a promising subject for researchers, for its great potential in overcoming the frustrating status quo in clinic, promoting welfare of TBI patient.
Collapse
Affiliation(s)
- Yibin Jia
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Guanyi Wang
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Yuqing Ye
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China.,Department of Neurosurgery, PLA 163rd Hospital (Second Affiliated Hospital of Hunan Normal University), Changsha 410000, China
| | - Enming Kang
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Huijun Chen
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Zishuo Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China
| | - Xiaosheng He
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China
| |
Collapse
|
30
|
Lengel D, Sevilla C, Romm ZL, Huh JW, Raghupathi R. Stem Cell Therapy for Pediatric Traumatic Brain Injury. Front Neurol 2020; 11:601286. [PMID: 33343501 PMCID: PMC7738475 DOI: 10.3389/fneur.2020.601286] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022] Open
Abstract
There has been a growing interest in the potential of stem cell transplantation as therapy for pediatric brain injuries. Studies in pre-clinical models of pediatric brain injury such as Traumatic Brain Injury (TBI) and neonatal hypoxia-ischemia (HI) have contributed to our understanding of the roles of endogenous stem cells in repair processes and functional recovery following brain injury, and the effects of exogenous stem cell transplantation on recovery from brain injury. Although only a handful of studies have evaluated these effects in models of pediatric TBI, many studies have evaluated stem cell transplantation therapy in models of neonatal HI which has a considerable overlap of injury pathology with pediatric TBI. In this review, we have summarized data on the effects of stem cell treatments on histopathological and functional outcomes in models of pediatric brain injury. Importantly, we have outlined evidence supporting the potential for stem cell transplantation to mitigate pathology of pediatric TBI including neuroinflammation and white matter injury, and challenges that will need to be addressed to incorporate these therapies to improve functional outcomes following pediatric TBI.
Collapse
Affiliation(s)
- Dana Lengel
- Graduate Program in Neuroscience, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Cruz Sevilla
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Zoe L Romm
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Jimmy W Huh
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Ramesh Raghupathi
- Graduate Program in Neuroscience, Drexel University College of Medicine, Philadelphia, PA, United States.,Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
31
|
Enam SF, Kader SR, Bodkin N, Lyon JG, Calhoun M, Azrak C, Tiwari PM, Vanover D, Wang H, Santangelo PJ, Bellamkonda RV. Evaluation of M2-like macrophage enrichment after diffuse traumatic brain injury through transient interleukin-4 expression from engineered mesenchymal stromal cells. J Neuroinflammation 2020; 17:197. [PMID: 32563258 PMCID: PMC7306141 DOI: 10.1186/s12974-020-01860-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/29/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Appropriately modulating inflammation after traumatic brain injury (TBI) may prevent disabilities for the millions of those inflicted annually. In TBI, cellular mediators of inflammation, including macrophages and microglia, possess a range of phenotypes relevant for an immunomodulatory therapeutic approach. It is thought that early phenotypic modulation of these cells will have a cascading healing effect. In fact, an anti-inflammatory, "M2-like" macrophage phenotype after TBI has been associated with neurogenesis, axonal regeneration, and improved white matter integrity (WMI). There already exist clinical trials seeking an M2-like bias through mesenchymal stem/stromal cells (MSCs). However, MSCs do not endogenously synthesize key signals that induce robust M2-like phenotypes such as interleukin-4 (IL-4). METHODS To enrich M2-like macrophages in a clinically relevant manner, we augmented MSCs with synthetic IL-4 mRNA to transiently express IL-4. These IL-4 expressing MSCs (IL-4 MSCs) were characterized for expression and functionality and then delivered in a modified mouse TBI model of closed head injury. Groups were assessed for functional deficits and MR imaging. Brain tissue was analyzed through flow cytometry, multi-plex ELISA, qPCR, histology, and RNA sequencing. RESULTS We observed that IL-4 MSCs indeed induce a robust M2-like macrophage phenotype and promote anti-inflammatory gene expression after TBI. However, here we demonstrate that acute enrichment of M2-like macrophages did not translate to improved functional or histological outcomes, or improvements in WMI on MR imaging. To further understand whether dysfunctional pathways underlie the lack of therapeutic effect, we report transcriptomic analysis of injured and treated brains. Through this, we discovered that inflammation persists despite acute enrichment of M2-like macrophages in the brain. CONCLUSION The results demonstrate that MSCs can be engineered to induce a stronger M2-like macrophage response in vivo. However, they also suggest that acute enrichment of only M2-like macrophages after diffuse TBI cannot orchestrate neurogenesis, axonal regeneration, or improve WMI. Here, we also discuss our modified TBI model and methods to assess severity, behavioral studies, and propose that IL-4 expressing MSCs may also have relevance in other cavitary diseases or in improving biomaterial integration into tissues.
Collapse
Affiliation(s)
- Syed Faaiz Enam
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Nicholas Bodkin
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Johnathan G Lyon
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Mark Calhoun
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Cesar Azrak
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Pooja Munnilal Tiwari
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Daryll Vanover
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Haichen Wang
- Department of Neurology, Duke University, Durham, NC, USA
| | - Philip J Santangelo
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | | |
Collapse
|
32
|
Hu Z, Gajavelli S, Spurlock MS, Mahavadi A, Quesada LS, Gajavelli GR, Andreoni CB, Di L, Janecki J, Lee SW, Rivera KN, Shear DA, Bullock RM. Human neural stem cell transplant location-dependent neuroprotection and motor deficit amelioration in rats with penetrating traumatic brain injury. J Trauma Acute Care Surg 2020; 88:477-485. [PMID: 31626023 PMCID: PMC7098436 DOI: 10.1097/ta.0000000000002510] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/30/2019] [Accepted: 09/17/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND Penetrating traumatic brain injury induces chronic inflammation that drives persistent tissue loss long after injury. Absence of endogenous reparative neurogenesis and effective neuroprotective therapies render injury-induced disability an unmet need. Cell replacement via neural stem cell transplantation could potentially rebuild the tissue and alleviate penetrating traumatic brain injury disability. The optimal transplant location remains to be determined. METHODS To test if subacute human neural stem cell (hNSC) transplant location influences engraftment, lesion expansion, and motor deficits, rats (n = 10/group) were randomized to the following four groups (uninjured and three injured): group 1 (Gr1), uninjured with cell transplants (sham+hNSCs), 1-week postunilateral penetrating traumatic brain injury, after establishing motor deficit; group 2 (Gr2), treated with vehicle (media, no cells); group 3 (Gr3), hNSCs transplanted into lesion core (intra); and group 4 (Gr4), hNSCs transplanted into tissue surrounding the lesion (peri). All animals were immunosuppressed for 12 weeks and euthanized following motor assessment. RESULTS In Gr2, penetrating traumatic brain injury effect manifests as porencephalic cyst, 22.53 ± 2.87 (% of intact hemisphere), with p value of <0.0001 compared with uninjured Gr1. Group 3 lesion volume at 17.44 ± 2.11 did not differ significantly from Gr2 (p = 0.36), while Gr4 value, 9.17 ± 1.53, differed significantly (p = 0.0001). Engraftment and neuronal differentiation were significantly lower in the uninjured Gr1 (p < 0.05), compared with injured groups. However, there were no differences between Gr3 and Gr4. Significant increase in cortical tissue sparing (p = 0.03), including motor cortex (p = 0.005) was observed in Gr4 but not Gr3. Presence of transplant within lesion or in penumbra attenuated motor deficit development (p < 0.05) compared with Gr2. CONCLUSION In aggregate, injury milieu supports transplanted cell proliferation and differentiation independent of location. Unexpectedly, cortical sparing is transplant location dependent. Thus, apart from cell replacement and transplant mediated deficit amelioration, transplant location-dependent neuroprotection may be key to delaying onset or preventing development of injury-induced disability. LEVEL OF EVIDENCE Preclinical study evaluation of therapeutic intervention, level VI.
Collapse
Affiliation(s)
- Zhen Hu
- From the Department of Neurosurgery (Z.H.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China; Miami Project to Cure Paralysis (Z.H., S.G., M.S.S., A.M., L.S.Q., G.R.G., C.B.A., L.D., J.J., S.W.L., K.N.R., R.M.D.), University of Miami, Miami, Florida; and Branch of Brain Trauma Neuroprotection and Neurorestoration (D.A.S.), Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Therajaran P, Hamilton JA, O'Brien TJ, Jones NC, Ali I. Microglial polarization in posttraumatic epilepsy: Potential mechanism and treatment opportunity. Epilepsia 2020; 61:203-215. [PMID: 31943156 DOI: 10.1111/epi.16424] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 12/17/2022]
Abstract
Owing to the complexity of the pathophysiological mechanisms driving epileptogenesis following traumatic brain injury (TBI), effective preventive treatment approaches are not yet available for posttraumatic epilepsy (PTE). Neuroinflammation appears to play a critical role in the pathogenesis of the acquired epilepsies, including PTE, but despite a large preclinical literature demonstrating the ability of anti-inflammatory treatments to suppress epileptogenesis and chronic seizures, no anti-inflammatory treatment approaches have been clinically proven to date. TBI triggers robust inflammatory cascades, suggesting that they may be relevant for the pathogenesis of PTE. A major cell type involved in such cascades is the microglial cells-brain-resident immune cells that become activated after brain injury. When activated, these cells can oscillate between different phenotypes, and such polarization states are associated with the release of various pro- and anti-inflammatory mediators that may influence brain repair processes, and also differentially contribute to the development of PTE. As the molecular mechanisms and key signaling molecules associated with microglial polarization in brain are discovered, strategies are now emerging that can modulate this polarization, promoting this as a potential therapeutic strategy for PTE. In this review, we discuss the relevant literature regarding the polarization of brain-resident immune cells following TBI and attempt to put into perspective a role in epilepsy pathogenesis. Finally, we explore potential strategies that could polarize microglia/macrophages toward a neuroprotective phenotype to mitigate PTE development.
Collapse
Affiliation(s)
- Peravina Therajaran
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - John A Hamilton
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Terence J O'Brien
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Nigel C Jones
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Idrish Ali
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
34
|
Nguyen H, Zarriello S, Coats A, Nelson C, Kingsbury C, Gorsky A, Rajani M, Neal EG, Borlongan CV. Stem cell therapy for neurological disorders: A focus on aging. Neurobiol Dis 2019; 126:85-104. [PMID: 30219376 PMCID: PMC6650276 DOI: 10.1016/j.nbd.2018.09.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/04/2018] [Accepted: 09/11/2018] [Indexed: 02/07/2023] Open
Abstract
Age-related neurological disorders continue to pose a significant societal and economic burden. Aging is a complex phenomenon that affects many aspects of the human body. Specifically, aging can have detrimental effects on the progression of brain diseases and endogenous stem cells. Stem cell therapies possess promising potential to mitigate the neurological symptoms of such diseases. However, aging presents a major obstacle for maximum efficacy of these treatments. In this review, we discuss current preclinical and clinical literature to highlight the interactions between aging, stem cell therapy, and the progression of major neurological disease states such as Parkinson's disease, Huntington's disease, stroke, traumatic brain injury, amyotrophic lateral sclerosis, multiple sclerosis, and multiple system atrophy. We raise important questions to guide future research and advance novel treatment options.
Collapse
Affiliation(s)
- Hung Nguyen
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Sydney Zarriello
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Alexandreya Coats
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Cannon Nelson
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Chase Kingsbury
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Anna Gorsky
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Mira Rajani
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Elliot G Neal
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA.
| |
Collapse
|
35
|
Kassi AAY, Mahavadi AK, Clavijo A, Caliz D, Lee SW, Ahmed AI, Yokobori S, Hu Z, Spurlock MS, Wasserman JM, Rivera KN, Nodal S, Powell HR, Di L, Torres R, Leung LY, Rubiano AM, Bullock RM, Gajavelli S. Enduring Neuroprotective Effect of Subacute Neural Stem Cell Transplantation After Penetrating TBI. Front Neurol 2019; 9:1097. [PMID: 30719019 PMCID: PMC6348935 DOI: 10.3389/fneur.2018.01097] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 12/03/2018] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury (TBI) is the largest cause of death and disability of persons under 45 years old, worldwide. Independent of the distribution, outcomes such as disability are associated with huge societal costs. The heterogeneity of TBI and its complicated biological response have helped clarify the limitations of current pharmacological approaches to TBI management. Five decades of effort have made some strides in reducing TBI mortality but little progress has been made to mitigate TBI-induced disability. Lessons learned from the failure of numerous randomized clinical trials and the inability to scale up results from single center clinical trials with neuroprotective agents led to the formation of organizations such as the Neurological Emergencies Treatment Trials (NETT) Network, and international collaborative comparative effectiveness research (CER) to re-orient TBI clinical research. With initiatives such as TRACK-TBI, generating rich and comprehensive human datasets with demographic, clinical, genomic, proteomic, imaging, and detailed outcome data across multiple time points has become the focus of the field in the United States (US). In addition, government institutions such as the US Department of Defense are investing in groups such as Operation Brain Trauma Therapy (OBTT), a multicenter, pre-clinical drug-screening consortium to address the barriers in translation. The consensus from such efforts including "The Lancet Neurology Commission" and current literature is that unmitigated cell death processes, incomplete debris clearance, aberrant neurotoxic immune, and glia cell response induce progressive tissue loss and spatiotemporal magnification of primary TBI. Our analysis suggests that the focus of neuroprotection research needs to shift from protecting dying and injured neurons at acute time points to modulating the aberrant glial response in sub-acute and chronic time points. One unexpected agent with neuroprotective properties that shows promise is transplantation of neural stem cells. In this review we present (i) a short survey of TBI epidemiology and summary of current care, (ii) findings of past neuroprotective clinical trials and possible reasons for failure based upon insights from human and preclinical TBI pathophysiology studies, including our group's inflammation-centered approach, (iii) the unmet need of TBI and unproven treatments and lastly, (iv) present evidence to support the rationale for sub-acute neural stem cell therapy to mediate enduring neuroprotection.
Collapse
Affiliation(s)
- Anelia A. Y. Kassi
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Anil K. Mahavadi
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Angelica Clavijo
- Neurosurgery Service, INUB-MEDITECH Research Group, El Bosque University, Bogotá, CO, United States
| | - Daniela Caliz
- Neurosurgery Service, INUB-MEDITECH Research Group, El Bosque University, Bogotá, CO, United States
| | - Stephanie W. Lee
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Aminul I. Ahmed
- Wessex Neurological Centre, University Hospitals Southampton, Southampton, United Kingdom
| | - Shoji Yokobori
- Department of Emergency and Critical Care Medicine, Nippon Medical School, Tokyo, Japan
| | - Zhen Hu
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Markus S. Spurlock
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Joseph M Wasserman
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Karla N. Rivera
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Samuel Nodal
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Henry R. Powell
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Long Di
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Rolando Torres
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Lai Yee Leung
- Branch of Brain Trauma Neuroprotection and Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Andres Mariano Rubiano
- Neurosurgery Service, INUB-MEDITECH Research Group, El Bosque University, Bogotá, CO, United States
| | - Ross M. Bullock
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Shyam Gajavelli
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
36
|
Chen D, Hu S, Liu J, Li S. E-cadherin regulates biological behaviors of neural stem cells and promotes motor function recovery following spinal cord injury. Exp Ther Med 2019; 17:2061-2070. [PMID: 30783478 PMCID: PMC6364216 DOI: 10.3892/etm.2019.7176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 11/26/2018] [Indexed: 02/06/2023] Open
Abstract
Stem cell-based repair strategies for spinal cord injury (SCI) are a highly studied area of research. Multiple gene-modified stem cells have been transplanted into SCI models, in the hope of generating more neurons to repair a damaged nervous system. However, the results are not always successful, as the grafted cells may be unable to survive in the injured spinal cord. E-cadherin, a transmembrane adhesion protein, has been identified as an epithelial-to-mesenchymal transition marker and is vital for morphological structure maintenance and the functional integrity of epithelial cells. At present, few studies have examined the association between E-cadherin and neural stem cells (NSCs). The present study investigated the expression of E-cadherin in subcultured NSCs and differentiated NSCs. Furthermore, the effect of E-cadherin on NSC viability, migration, differentiation and neurosphere formation was assessed. An in vivo study was used to assess the long-term survival of grafted NSCs. Additionally, the protective effect of E-cadherin on SCI was assessed by analyzing tissue repair, Basso Mouse Scale scores and the expression of inflammatory cytokines. The results of the present study suggested that E-cadherin was able to promote NSC viability and neurosphere formation; however, it had no significant effect on NSC differentiation. To conclude, grafted NSCs with highly expressed E-cadherin facilitated motor function recovery following SCI by reducing the release of inflammatory cytokines.
Collapse
Affiliation(s)
- Dong Chen
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Siyuan Hu
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Jie Liu
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Shaohua Li
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| |
Collapse
|
37
|
Silveira GDP, Ishimura ME, Teixeira D, Galindo LT, Sardinha AA, Porcionatto M, Longo-Maugéri IM. Improvement of Mesenchymal Stem Cell Immunomodulatory Properties by Heat-Killed Propionibacterium acnes via TLR2. Front Mol Neurosci 2019; 11:489. [PMID: 30687005 PMCID: PMC6336115 DOI: 10.3389/fnmol.2018.00489] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 12/14/2018] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are an essential tool for regenerative medicine, which aims to develop new technologies to improve their effects to obtain useful transplantation results. MSC immunomodulatory role has been just demonstrated; however, how they react when they are stimulated by an adjuvant is poorly understood. Our group showed the adjuvant effect of killed Propionibacterium acnes (P. acnes) on hematopoietic stem cells. As these cells share the same MSCs bone marrow (BM) site and interact with each other, here we evaluated the P. acnes and its soluble polysaccharide (PS) effect on MSCs and their immunomodulatory role in a murine model of traumatic brain injury (TBI). The bacteria increased the absolute number of MSCs, including MSC subpopulations, and maintained MSC plasticity. P. acnes and PS enhanced MSC proliferation and improved their immunomodulatory effect. P. acnes-MSC and PS-MSC transplantation increased anti-inflammatory cytokine expression and diminished pro-inflammatory cytokine expression after injury. This effect seemed to be mediated via TLR2 since P. acnes-KOTLR2-MSC transplantation decreased TGF-β and IL-10 expression. Increasing in neural stem cells and neuroblasts after PS-MSC transplantation was also observed. The adjuvant effect of P. acnes is an alternative means of expanding MSCs and important to identify their subpopulations to know better their role under exogenous stimuli including inflammation resolution in an experimental model.
Collapse
Affiliation(s)
- Gabriela da Paz Silveira
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Mayari Eika Ishimura
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Daniela Teixeira
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Layla Tesla Galindo
- Division of Molecular Biology, Department of Biochemistry, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Agnes Araujo Sardinha
- Division of Molecular Biology, Department of Biochemistry, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Marimelia Porcionatto
- Division of Molecular Biology, Department of Biochemistry, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Ieda Maria Longo-Maugéri
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
38
|
Zibara K, Ballout N, Mondello S, Karnib N, Ramadan N, Omais S, Nabbouh A, Caliz D, Clavijo A, Hu Z, Ghanem N, Gajavelli S, Kobeissy F. Combination of drug and stem cells neurotherapy: Potential interventions in neurotrauma and traumatic brain injury. Neuropharmacology 2018; 145:177-198. [PMID: 30267729 DOI: 10.1016/j.neuropharm.2018.09.032] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 09/17/2018] [Accepted: 09/21/2018] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury (TBI) has been recognized as one of the major public health issues that leads to devastating neurological disability. As a consequence of primary and secondary injury phases, neuronal loss following brain trauma leads to pathophysiological alterations on the molecular and cellular levels that severely impact the neuropsycho-behavioral and motor outcomes. Thus, to mitigate the neuropathological sequelae post-TBI such as cerebral edema, inflammation and neural degeneration, several neurotherapeutic options have been investigated including drug intervention, stem cell use and combinational therapies. These treatments aim to ameliorate cellular degeneration, motor decline, cognitive and behavioral deficits. Recently, the use of neural stem cells (NSCs) coupled with selective drug therapy has emerged as an alternative treatment option for neural regeneration and behavioral rehabilitation post-neural injury. Given their neuroprotective abilities, NSC-based neurotherapy has been widely investigated and well-reported in numerous disease models, notably in trauma studies. In this review, we will elaborate on current updates in cell replacement therapy in the area of neurotrauma. In addition, we will discuss novel combination drug therapy treatments that have been investigated in conjunction with stem cells to overcome the limitations associated with stem cell transplantation. Understanding the regenerative capacities of stem cell and drug combination therapy will help improve functional recovery and brain repair post-TBI. This article is part of the Special Issue entitled "Novel Treatments for Traumatic Brain Injury".
Collapse
Affiliation(s)
- Kazem Zibara
- ER045, Laboratory of Stem Cells, PRASE, Lebanese University, Beirut, Lebanon; Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Nissrine Ballout
- ER045, Laboratory of Stem Cells, PRASE, Lebanese University, Beirut, Lebanon
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Nabil Karnib
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon
| | - Naify Ramadan
- Department of Women's and Children's Health (KBH), Division of Clinical Pediatrics, Karolinska Institute, Sweden
| | - Saad Omais
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Ali Nabbouh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon
| | - Daniela Caliz
- Lois Pope LIFE Center, Neurosurgery, University of Miami, 33136, Miami, FL, USA
| | - Angelica Clavijo
- Lois Pope LIFE Center, Neurosurgery, University of Miami, 33136, Miami, FL, USA
| | - Zhen Hu
- Lois Pope LIFE Center, Neurosurgery, University of Miami, 33136, Miami, FL, USA
| | - Noël Ghanem
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Shyam Gajavelli
- Lois Pope LIFE Center, Neurosurgery, University of Miami, 33136, Miami, FL, USA.
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon; Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Department of Emergency Medicine, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
39
|
Pearse DD, Bastidas J, Izabel SS, Ghosh M. Schwann Cell Transplantation Subdues the Pro-Inflammatory Innate Immune Cell Response after Spinal Cord Injury. Int J Mol Sci 2018; 19:E2550. [PMID: 30154346 PMCID: PMC6163303 DOI: 10.3390/ijms19092550] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 08/20/2018] [Accepted: 08/22/2018] [Indexed: 12/12/2022] Open
Abstract
The transplantation of Schwann cells (SCs) has been shown to provide tissue preservation and support axon growth and remyelination as well as improve functional recovery across a diverse range of experimental spinal cord injury (SCI) paradigms. The autologous use of SCs has progressed to Phase 1 SCI clinical trials in humans where their use has been shown to be both feasible and safe. The contribution of immune modulation to the protective and reparative actions of SCs within the injured spinal cord remains largely unknown. In the current investigation, the ability of SC transplants to alter the innate immune response after contusive SCI in the rat was examined. SCs were intraspinally transplanted into the lesion site at 1 week following a thoracic (T8) contusive SCI. Multicolor flow cytometry and immunohistochemical analysis of specific phenotypic markers of pro- and anti-inflammatory microglia and macrophages as well as cytokines at 1 week after SC transplantation was employed. The introduction of SCs significantly attenuated the numbers of cluster of differentiation molecule 11B (CD11b)⁺, cluster of differentiation molecule 68 (CD68)⁺, and ionized calcium-binding adapter molecule 1 (Iba1)⁺ immune cells within the lesion implant site, particularly those immunoreactive for the pro-inflammatory marker, inducible nitric oxide synthase (iNOS). Whereas numbers of anti-inflammatory CD68⁺ Arginase-1 (Arg1⁺) iNOS- cells were not altered by SC transplantation, CD68⁺ cells of an intermediate, Arg1⁺ iNOS⁺ phenotype were increased by the introduction of SCs into the injured spinal cord. The morphology of Iba1⁺ immune cells was also markedly altered in the SC implant, being elongated and in alignment with SCs and in-growing axons versus their amoeboid form after SCI alone. Examination of pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), and anti-inflammatory cytokines, interleukin-4 (IL-4) and interleukin-10 (IL-10), by multicolor flow cytometry analysis showed that their production in CD11b⁺ cells was unaltered by SC transplantation at 1 week post-transplantation. The ability of SCs to subdue the pro-inflammatory iNOS⁺ microglia and macrophage phenotype after intraspinal transplantation may provide an important contribution to the neuroprotective effects of SCs within the sub-acute SCI setting.
Collapse
Affiliation(s)
- Damien D Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA.
| | - Johana Bastidas
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Sarah S Izabel
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Mousumi Ghosh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA.
| |
Collapse
|
40
|
Ali I, Silva JC, Liu S, Shultz SR, Kwan P, Jones NC, O'Brien TJ. Targeting neurodegeneration to prevent post-traumatic epilepsy. Neurobiol Dis 2018; 123:100-109. [PMID: 30099094 DOI: 10.1016/j.nbd.2018.08.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/31/2018] [Accepted: 08/08/2018] [Indexed: 12/14/2022] Open
Abstract
In the quest for developing new therapeutic targets for post-traumatic epilepsies (PTE), identifying mechanisms relevant to development and progression of disease is critical. A growing body of literature suggests involvement of neurodegenerative mechanisms in the pathophysiology of acquired epilepsies, including following traumatic brain injury (TBI). In this review, we discuss the potential of some of these mechanisms to be targets for the development of a therapy against PTE.
Collapse
Affiliation(s)
- Idrish Ali
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Melbourne, Australia
| | - Juliana C Silva
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Melbourne, Australia
| | - Shijie Liu
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Melbourne, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Melbourne, Australia
| | - Patrick Kwan
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Melbourne, Australia
| | - Nigel C Jones
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Melbourne, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
41
|
Neal EG, Liska MG, Lippert T, Lin R, Gonzalez M, Russo E, Xu K, Ji X, Vale FL, Van Loveren H, Borlongan CV. An update on intracerebral stem cell grafts. Expert Rev Neurother 2018; 18:557-572. [PMID: 29961357 DOI: 10.1080/14737175.2018.1491309] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Primary neurological disorders are notoriously debilitating and deadly, and over the past four decades stem cell therapy has emerged as a promising treatment. Translation of stem cell therapies from the bench to the clinic requires a better understanding of delivery protocols, safety profile, and efficacy in each disease. Areas covered: In this review, benefits and risks of intracerebral stem cell transplantation are presented for consideration. Milestone discoveries in stem cell applications are reviewed to examine the efficacy and safety of intracerebral stem cell transplant therapy for disorders of the central nervous system and inform design of translatable protocols for clinically feasible stem cell-based treatments. Expert commentary: Intracerebral administration, compared to peripheral delivery, is more invasive and carries the risk of open brain surgery. However, direct cell implantation bypasses the blood-brain barrier and reduces the first-pass effect, effectively increasing the therapeutic cell deposition at its intended site of action. These benefits must be weighed with the risk of graft-versus-host immune response. Rigorous clinical trials are underway to assess the safety and efficacy of intracerebral transplants, and if successful will lead to widely available stem cell therapies for neurologic diseases in the coming years.
Collapse
Affiliation(s)
- Elliot G Neal
- a Department of Neurosurgery and Brain Repair , Center of Excellence for Aging and Brain Repair, USF Morsani College of Medicine , Tampa , FL , USA
| | - M Grant Liska
- a Department of Neurosurgery and Brain Repair , Center of Excellence for Aging and Brain Repair, USF Morsani College of Medicine , Tampa , FL , USA
| | - Trenton Lippert
- a Department of Neurosurgery and Brain Repair , Center of Excellence for Aging and Brain Repair, USF Morsani College of Medicine , Tampa , FL , USA
| | - Roger Lin
- a Department of Neurosurgery and Brain Repair , Center of Excellence for Aging and Brain Repair, USF Morsani College of Medicine , Tampa , FL , USA
| | - Melissa Gonzalez
- a Department of Neurosurgery and Brain Repair , Center of Excellence for Aging and Brain Repair, USF Morsani College of Medicine , Tampa , FL , USA
| | - Eleonora Russo
- a Department of Neurosurgery and Brain Repair , Center of Excellence for Aging and Brain Repair, USF Morsani College of Medicine , Tampa , FL , USA
| | - Kaya Xu
- a Department of Neurosurgery and Brain Repair , Center of Excellence for Aging and Brain Repair, USF Morsani College of Medicine , Tampa , FL , USA
| | - Xunming Ji
- b Department of Neurosurgery , Xuanwu Hospital, Capital Medical University , Beijing , China
| | - Fernando L Vale
- c USF Department of Neurosurgery and Brain Repair , Tampa , FL , USA
| | - Harry Van Loveren
- c USF Department of Neurosurgery and Brain Repair , Tampa , FL , USA
| | - Cesario V Borlongan
- a Department of Neurosurgery and Brain Repair , Center of Excellence for Aging and Brain Repair, USF Morsani College of Medicine , Tampa , FL , USA
| |
Collapse
|
42
|
Gincberg G, Shohami E, Trembovler V, Alexandrovich AG, Lazarovici P, Elchalal U. Nerve growth factor plays a role in the neurotherapeutic effect of a CD45 + pan-hematopoietic subpopulation derived from human umbilical cord blood in a traumatic brain injury model. Cytotherapy 2017; 20:245-261. [PMID: 29274773 DOI: 10.1016/j.jcyt.2017.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 10/09/2017] [Accepted: 11/14/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND AIMS Human umbilical cord blood (HUCB) is an important source of stem cells for therapy of hematopoietic disorders and is a potential therapy for various neurological disorders, including traumatic brain injury (TBI). The expression of nerve growth factor (NGF) and its receptors TrkA, p75NTR and α9β1 integrin on an HUCB CD45+ pan-hematopoietic subpopulation was investigated in the context of its neurotherapeutic potential after TBI. METHODS NGF and its receptors were detected on CD45+ cells by reverse transcriptase polymerase chain reaction, flow cytometry analysis and confocal microscopy. CD45+ cells were stimulated by TBI brain extracts, and NGF levels were measured by enzyme-linked immunosorbent assay. TBI mice were divided into six groups for xenogeneic intravenous transplantation, 1 day post-trauma, with 1 × 106 CD45+ cells untreated or treated with the anti-NGF neutralizing antibody K252a, a TrkA antagonist; VLO5, an α9β1 disintegrin; or negative (vehicle) and positive (NGF) controls. RESULTS The HUCB CD45+ subpopulation constitutively expresses NGF and its receptors, mainly TrkA and p75NTR and minor levels of α9β1. In vitro experiments provided evidence that trauma-related mediators from brain extracts of TBI mice induced release of NGF from HUCB CD45+ cell cultures. HUCB CD45+ cells induced a neurotherapeutic effect in TBI mice, abrogated by cell treatment with either anti-NGF antibody or K252a, but not VLO5. CONCLUSIONS These findings strengthen the role of NGF and its TrkA receptor in the HUCB CD45+ subpopulation's neurotherapeutic effect. The presence of neurotrophin receptors in the HUCB CD45+ pan-hematopoietic subpopulation may explain the neuroprotective effect of cord blood in therapy of a variety of neurological disorders.
Collapse
Affiliation(s)
- Galit Gincberg
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Esther Shohami
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Victoria Trembovler
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Alexander G Alexandrovich
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Uriel Elchalal
- Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
43
|
Galgano M, Toshkezi G, Qiu X, Russell T, Chin L, Zhao LR. Traumatic Brain Injury: Current Treatment Strategies and Future Endeavors. Cell Transplant 2017; 26:1118-1130. [PMID: 28933211 PMCID: PMC5657730 DOI: 10.1177/0963689717714102] [Citation(s) in RCA: 319] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 10/16/2016] [Accepted: 10/18/2016] [Indexed: 01/04/2023] Open
Abstract
Traumatic brain injury (TBI) presents in various forms ranging from mild alterations of consciousness to an unrelenting comatose state and death. In the most severe form of TBI, the entirety of the brain is affected by a diffuse type of injury and swelling. Treatment modalities vary extensively based on the severity of the injury and range from daily cognitive therapy sessions to radical surgery such as bilateral decompressive craniectomies. Guidelines have been set forth regarding the optimal management of TBI, but they must be taken in context of the situation and cannot be used in every individual circumstance. In this review article, we have summarized the current status of treatment for TBI in both clinical practice and basic research. We have put forth a brief overview of the various subtypes of traumatic injuries, optimal medical management, and both the noninvasive and invasive monitoring modalities, in addition to the surgical interventions necessary in particular instances. We have overviewed the main achievements in searching for therapeutic strategies of TBI in basic science. We have also discussed the future direction for developing TBI treatment from an experimental perspective.
Collapse
Affiliation(s)
- Michael Galgano
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Gentian Toshkezi
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Xuecheng Qiu
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
- VA Health Care Upstate New York, Syracuse VA Medical Center, Syracuse, NY, USA
| | - Thomas Russell
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Lawrence Chin
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Li-Ru Zhao
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, NY, USA
- VA Health Care Upstate New York, Syracuse VA Medical Center, Syracuse, NY, USA
| |
Collapse
|
44
|
Spurlock MS, Ahmed AI, Rivera KN, Yokobori S, Lee SW, Sam PN, Shear DA, Hefferan MP, Hazel TG, Johe KK, Gajavelli S, Tortella FC, Bullock RM. Amelioration of Penetrating Ballistic-Like Brain Injury Induced Cognitive Deficits after Neuronal Differentiation of Transplanted Human Neural Stem Cells. J Neurotrauma 2017; 34:1981-1995. [PMID: 28249550 PMCID: PMC6913783 DOI: 10.1089/neu.2016.4602] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Penetrating traumatic brain injury (PTBI) is one of the major cause of death and disability worldwide. Previous studies with penetrating ballistic-like brain injury (PBBI), a PTBI rat model revealed widespread perilesional neurodegeneration, similar to that seen in humans following gunshot wound to the head, which is unmitigated by any available therapies to date. Therefore, we evaluated human neural stem cell (hNSC) engraftment to putatively exploit the potential of cell therapy that has been seen in other central nervous system injury models. Toward this objective, green fluorescent protein (GFP) labeled hNSC (400,000 per animal) were transplanted in immunosuppressed Sprague-Dawley (SD), Fisher, and athymic (ATN) PBBI rats 1 week after injury. Tacrolimus (3 mg/kg 2 days prior to transplantation, then 1 mg/kg/day), methylprednisolone (10 mg/kg on the day of transplant, 1 mg/kg/week thereafter), and mycophenolate mofetil (30 mg/kg/day) for 7 days following transplantation were used to confer immunosuppression. Engraftment in SD and ATN was comparable at 8 weeks post-transplantation. Evaluation of hNSC differentiation and distribution revealed increased neuronal differentiation of transplanted cells with time. At 16 weeks post-transplantation, neither cell proliferation nor glial lineage markers were detected. Transplanted cell morphology was similar to that of neighboring host neurons, and there was relatively little migration of cells from the peritransplant site. By 16 weeks, GFP-positive processes extended both rostrocaudally and bilaterally into parenchyma, spreading along host white matter tracts, traversing the internal capsule, and extending ∼13 mm caudally from transplantation site reaching into the brainstem. In a Morris water maze test at 8 weeks post-transplantation, animals with transplants had shorter latency to platform than vehicle-treated animals. However, weak injury-induced cognitive deficits in the control group at the delayed time point confounded benefits of durable engraftment and neuronal differentiation. Therefore, these results justify further studies to progress towards clinical translation of hNSC therapy for PTBI.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Deborah A. Shear
- Branch of Brain Trauma Neuroprotection and Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | | | | | | | | | - Frank C. Tortella
- Branch of Brain Trauma Neuroprotection and Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | | |
Collapse
|
45
|
Michels M, Sonai B, Dal-Pizzol F. Polarization of microglia and its role in bacterial sepsis. J Neuroimmunol 2017; 303:90-98. [PMID: 28087076 DOI: 10.1016/j.jneuroim.2016.12.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/04/2016] [Accepted: 12/28/2016] [Indexed: 12/14/2022]
Abstract
Microglial polarization in response to brain inflammatory conditions is a crescent field in neuroscience. However, the effect of systemic inflammation, and specifically sepsis, is a relatively unexplored field that has great interest and relevance. Sepsis has been associated with both early and late harmful events of the central nervous system, suggesting that there is a close link between sepsis and neuroinflammation. During sepsis evolution it is supposed that microglial could exert both neurotoxic and repairing effects depending on the specific microglial phenotype assumed. In this context, here it was reviewed the role of microglial polarization during sepsis-associated brain dysfunction.
Collapse
Affiliation(s)
- Monique Michels
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Av Universitária, 1105, Criciúma 88806000, SC, Brazil.
| | - Beatriz Sonai
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Av Universitária, 1105, Criciúma 88806000, SC, Brazil.
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Av Universitária, 1105, Criciúma 88806000, SC, Brazil; Center of Excellence in Applied Neurosciences of Santa Catarina (NENASC), Graduate Program in Medical Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil.
| |
Collapse
|
46
|
The Polarization States of Microglia in TBI: A New Paradigm for Pharmacological Intervention. Neural Plast 2017; 2017:5405104. [PMID: 28255460 PMCID: PMC5309408 DOI: 10.1155/2017/5405104] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/17/2016] [Accepted: 01/11/2017] [Indexed: 01/04/2023] Open
Abstract
Traumatic brain injury (TBI) is a serious medical and social problem worldwide. Because of the complex pathophysiological mechanisms of TBI, effective pharmacotherapy is still lacking. The microglial cells are resident tissue macrophages located in the brain and have two major polarization states, M1 phenotype and M2 phenotype, when activated. The M1 phenotype is related to the release of proinflammatory cytokines and secondary brain injury, while the M2 phenotype has been proved to be responsible for the release of anti-inflammation cytokines and for central nervous system (CNS) repair. In animal models, pharmacological strategies inhibiting the M1 phenotype and promoting the M2 phenotype of microglial cells could alleviate cerebral damage and improve neurological function recovery after TBI. In this review, we aimed to summarize the current knowledge about the pathological significance of microglial M1/M2 polarization in the pathophysiology of TBI. In addition, we reviewed several drugs that have provided neuroprotective effects against brain injury following TBI by altering the polarization states of the microglia. We emphasized that future investigation of the regulation mechanisms of microglial M1/M2 polarization in TBI is anticipated, which could contribute to the development of new targets of pharmacological intervention in TBI.
Collapse
|
47
|
Lee JH, Wei ZZ, Cao W, Won S, Gu X, Winter M, Dix TA, Wei L, Yu SP. Regulation of therapeutic hypothermia on inflammatory cytokines, microglia polarization, migration and functional recovery after ischemic stroke in mice. Neurobiol Dis 2016; 96:248-260. [PMID: 27659107 PMCID: PMC5161414 DOI: 10.1016/j.nbd.2016.09.013] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/30/2016] [Accepted: 09/17/2016] [Indexed: 01/08/2023] Open
Abstract
Stroke is a leading threat to human life and health in the US and around the globe, while very few effective treatments are available for stroke patients. Preclinical and clinical studies have shown that therapeutic hypothermia (TH) is a potential treatment for stroke. Using novel neurotensin receptor 1 (NTR1) agonists, we have demonstrated pharmacologically induced hypothermia and protective effects against brain damages after ischemic stroke, hemorrhage stroke, and traumatic brain injury (TBI) in rodent models. To further characterize the mechanism of TH-induced brain protection, we examined the effect of TH (at ±33°C for 6h) induced by the NTR1 agonist HPI-201 or physical (ice/cold air) cooling on inflammatory responses after ischemic stroke in mice and oxygen glucose deprivation (OGD) in cortical neuronal cultures. Seven days after focal cortical ischemia, microglia activation in the penumbra reached a peak level, which was significantly attenuated by TH treatments commenced 30min after stroke. The TH treatment decreased the expression of M1 type reactive factors including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-12, IL-23, and inducible nitric oxide synthase (iNOS) measured by RT-PCR and Western blot analyses. Meanwhile, TH treatments increased the expression of M2 type reactive factors including IL-10, Fizz1, Ym1, and arginase-1. In the ischemic brain and in cortical neuronal/BV2 microglia cultures subjected to OGD, TH attenuated the expression of monocyte chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein-1α (MIP-1α), two key chemokines in the regulation of microglia activation and infiltration. Consistently, physical cooling during OGD significantly decreased microglia migration 16h after OGD. Finally, TH improved functional recovery at 1, 3, and 7days after stroke. This study reveals the first evidence for hypothermia mediated regulation on inflammatory factor expression, microglia polarization, migration and indicates that the anti-inflammatory effect is an important mechanism underlying the brain protective effects of a TH therapy.
Collapse
Affiliation(s)
- Jin Hwan Lee
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, United States; Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, United States
| | - Zheng Z Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, United States; Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, United States
| | - Wenyuan Cao
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Soonmi Won
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, United States; Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, United States
| | - Megan Winter
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Thomas A Dix
- JT Pharmaceuticals, Mt. Pleasant, SC 29464, United States; Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29401, United States
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, United States; Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, United States; Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, United States.
| |
Collapse
|
48
|
Shen Y, Huang J, Liu L, Xu X, Han C, Zhang G, Jiang H, Li J, Lin Z, Xiong N, Wang T. A Compendium of Preparation and Application of Stem Cells in Parkinson's Disease: Current Status and Future Prospects. Front Aging Neurosci 2016; 8:117. [PMID: 27303288 PMCID: PMC4885841 DOI: 10.3389/fnagi.2016.00117] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/09/2016] [Indexed: 12/22/2022] Open
Abstract
Parkinson's Disease (PD) is a progressively neurodegenerative disorder, implicitly characterized by a stepwise loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) and explicitly marked by bradykinesia, rigidity, resting tremor and postural instability. Currently, therapeutic approaches available are mainly palliative strategies, including L-3,4-dihydroxy-phenylalanine (L-DOPA) replacement therapy, DA receptor agonist and deep brain stimulation (DBS) procedures. As the disease proceeds, however, the pharmacotherapeutic efficacy is inevitably worn off, worse still, implicated by side effects of motor response oscillations as well as L-DOPA induced dyskinesia (LID). Therefore, the frustrating status above has propeled the shift to cell replacement therapy (CRT), a promising restorative therapy intending to secure a long-lasting relief of patients' symptoms. By far, stem cell lines of multifarious origins have been established, which can be further categorized into embryonic stem cells (ESCs), neural stem cells (NSCs), induced neural stem cells (iNSCs), mesenchymal stem cells (MSCs), and induced pluripotent stem cells (iPSCs). In this review, we intend to present a compendium of preparation and application of multifarious stem cells, especially in relation to PD research and therapy. In addition, the current status, potential challenges and future prospects for practical CRT in PD patients will be elaborated as well.
Collapse
Affiliation(s)
- Yan Shen
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Jinsha Huang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Ling Liu
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Xiaoyun Xu
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Chao Han
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Guoxin Zhang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Haiyang Jiang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Jie Li
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Zhicheng Lin
- Department of Psychiatry, Harvard Medical School, Division of Alcohol and Drug Abuse, and Mailman Neuroscience Research Center, McLean Hospital Belmont, MA, USA
| | - Nian Xiong
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Tao Wang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| |
Collapse
|