1
|
Deng Q, Parker E, Duan R, Yang L. Preconditioning and Posttreatment Strategies in Neonatal Hypoxic-Ischemic Encephalopathy: Recent Advances and Clinical Challenges. Mol Neurobiol 2025:10.1007/s12035-025-04896-4. [PMID: 40178781 DOI: 10.1007/s12035-025-04896-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/24/2025] [Indexed: 04/05/2025]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is a severe neurological disorder caused by impaired cerebral blood flow and brain hypoxia, resulting in high morbidity and mortality rates. While therapeutic hypothermia remains the standard treatment and has been shown to reduce mortality to some extent, its therapeutic efficacy is limited, and it applies only to a select group of neonates who meet stringent inclusion criteria. Advances in our understanding of the pathophysiology of HIE have led to the identification of several promising neuroprotective strategies designed to mitigate or prevent the neurological damage induced by hypoxia-ischemia. Among these, preconditioning has emerged as a potent neuroprotective approach, enhancing cellular resilience to subsequent injury and potentially reducing treatment complexity and healthcare costs. Preconditioning/pretreatment and posttreatment offer significant promise in attenuating the neurological damage associated with HIE. Thus, exploring early intervention strategies for neonatal HIE, focusing on the comparative mechanisms and therapeutic targets of preconditioning and postconditioning, is critical to developing more effective treatment modalities. This review summarizes the current understanding of the pathophysiological mechanisms underlying neonatal HIE and its prevention and treatment strategies, providing new perspectives and a theoretical foundation for future neuroprotective interventions.
Collapse
Affiliation(s)
- Qianting Deng
- Laboratory of Exercise and Neurobiology, College of Physical Education and Sport Science, South China Normal University, Guangzhou, 510006, GD, China
| | - Emily Parker
- Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Rui Duan
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China.
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, College of Physical Education and Sport Science, South China Normal University, Guangzhou, 510006, GD, China.
| |
Collapse
|
2
|
Mintoft A, Vallatos A, Robertson NJ. Mesenchymal Stromal Cell therapy for Hypoxic Ischemic Encephalopathy: Future directions for combination therapy with hypothermia and/or melatonin. Semin Perinatol 2024; 48:151929. [PMID: 38902120 DOI: 10.1016/j.semperi.2024.151929] [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] [Indexed: 06/22/2024]
Abstract
Hypoxic ischemic encephalopathy (HIE) remains a leading cause of neonatal mortality and lifelong disability across the world. While therapeutic hypothermia (HT) is beneficial, it is only partially protective and adjuvant treatments that further improve outcomes are urgently needed. In high-income countries where HT is standard care, novel treatments are tested in conjunction with HT. Mesenchymal stromal cells (MSC) represent a paradigm shift in brain protection, uniquely adapting to the host cellular microenvironment. MSC have low immunogenicity and potent paracrine effects stimulating the host tissue repair and regeneration and reducing inflammation and apoptosis. Preclinical studies in perinatal brain injury suggest that MSC are beneficial after hypoxia-ischemia (HI) and most preclinical studies of MSC with HT show protection. Preclinical and early phase clinical trials have shown that allogenic administration of MSC to neonates with perinatal stroke and HIE is safe and feasible but further safety and efficacy studies of HT with MSC in these populations are needed. Combination therapies that target all stages of the evolution of injury after HI (eg HT, melatonin and MSC) show promise for improving outcomes in HIE.
Collapse
Affiliation(s)
- Alison Mintoft
- Institute for Women's Health, University College London, London, UK
| | - Antoine Vallatos
- School of Psychology and Neuroscience, University of Glasgow; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Nicola J Robertson
- Institute for Women's Health, University College London, London, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
3
|
Vaes JEG, Onstwedder SM, Trayford C, Gubbins E, Maas M, van Rijt SH, Nijboer CH. Modifying the Secretome of Mesenchymal Stem Cells Prolongs the Regenerative Treatment Window for Encephalopathy of Prematurity. Int J Mol Sci 2024; 25:6494. [PMID: 38928201 PMCID: PMC11203777 DOI: 10.3390/ijms25126494] [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: 02/15/2024] [Revised: 05/28/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Clinical treatment options to combat Encephalopathy of Prematurity (EoP) are still lacking. We, and others, have proposed (intranasal) mesenchymal stem cells (MSCs) as a potent therapeutic strategy to boost white matter repair in the injured preterm brain. Using a double-hit mouse model of diffuse white matter injury, we previously showed that the efficacy of MSC treatment was time dependent, with a significant decrease in functional and histological improvements after the postponement of cell administration. In this follow-up study, we aimed to investigate the mechanisms underlying this loss of therapeutic efficacy. Additionally, we optimized the regenerative potential of MSCs by means of genetic engineering with the transient hypersecretion of beneficial factors, in order to prolong the treatment window. Though the cerebral expression of known chemoattractants was stable over time, the migration of MSCs to the injured brain was partially impaired. Moreover, using a primary oligodendrocyte (OL) culture, we showed that the rescue of injured OLs was reduced after delayed MSC coculture. Cocultures of modified MSCs, hypersecreting IGF1, LIF, IL11, or IL10, with primary microglia and OLs, revealed a superior treatment efficacy over naïve MSCs. Additionally, we showed that the delayed intranasal administration of IGF1-, LIF-, or IL11-hypersecreting MSCs, improved myelination and the functional outcome in EoP mice. In conclusion, the impaired migration and regenerative capacity of intranasally applied MSCs likely underlie the observed loss of efficacy after delayed treatment. The intranasal administration of IGF1-, LIF-, or IL11-hypersecreting MSCs, is a promising optimization strategy to prolong the window for effective MSC treatment in preterm infants with EoP.
Collapse
Affiliation(s)
- Josine E. G. Vaes
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3508 AB Utrecht, The Netherlands
- Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3508 AB Utrecht, The Netherlands
| | - Suzanne M. Onstwedder
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3508 AB Utrecht, The Netherlands
| | - Chloe Trayford
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Eva Gubbins
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Mirjam Maas
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3508 AB Utrecht, The Netherlands
| | - Sabine H. van Rijt
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Cora H. Nijboer
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht University, 3508 AB Utrecht, The Netherlands
| |
Collapse
|
4
|
Milczarek O, Jarocha D, Starowicz-Filip A, Kasprzycki M, Kijowski J, Mordel A, Kwiatkowski S, Majka M. Bone Marrow Nucleated Cells and Bone Marrow-Derived CD271+ Mesenchymal Stem Cell in Treatment of Encephalopathy and Drug-Resistant Epilepsy. Stem Cell Rev Rep 2024; 20:1015-1025. [PMID: 38483743 DOI: 10.1007/s12015-023-10673-4] [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] [Accepted: 12/22/2023] [Indexed: 05/12/2024]
Abstract
The broad spectrum of brain injuries in preterm newborns and the plasticity of the central nervous system prompts us to seek solutions for neurodegeneration to prevent the consequences of prematurity and perinatal problems. The study aimed to evaluate the safety and efficacy of the implantation of autologous bone marrow nucleated cells and bone marrow mesenchymal stem cells in different schemes in patients with hypoxic-ischemic encephalopathy and immunological encephalopathy. Fourteen patients received single implantation of bone marrow nucleated cells administered intrathecally and intravenously, followed by multiple rounds of bone marrow mesenchymal stem cells implanted intrathecally, and five patients were treated only with repeated rounds of bone marrow mesenchymal stem cells. Seizure outcomes improved in most cases, including fewer seizures and status epilepticus and reduced doses of antiepileptic drugs compared to the period before treatment. The neuropsychological improvement was more frequent in patients with hypoxic-ischemic encephalopathy than in the immunological encephalopathy group. Changes in emotional functioning occurred with similar frequency in both groups of patients. In the hypoxic-ischemic encephalopathy group, motor improvement was observed in all patients and the majority in the immunological encephalopathy group. The treatment had manageable toxicity, mainly mild to moderate early-onset adverse events. The treatment was generally safe in the 4-year follow-up period, and the effects of the therapy were maintained after its termination.
Collapse
Affiliation(s)
- Olga Milczarek
- Faculty of Medicine, Department of Children's Neurosurgery, Jagiellonian University Medical College Institute of Pediatrics, Cracow, Poland.
| | - Danuta Jarocha
- Hematology Department, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anna Starowicz-Filip
- Faculty of Medicine, Department of Children's Neurosurgery, Jagiellonian University Medical College Institute of Pediatrics, Cracow, Poland
- Faculty of Medicine, Department of Psychology, Jagiellonian University Medicl College, Cracow, Poland
| | - Maciej Kasprzycki
- Students' Scientific Group at the Department of Pediatric Neurosurgery, Jagiellonian University Medical College Institute of Pediatrics, Cracow, Poland
| | - Jacek Kijowski
- Faculty of Medicine, Department of Transplantation, Jagiellonian University Medical College Institute of Pediatrics, Cracow, Poland
| | - Anna Mordel
- Faculty of Medicine, Department of Transplantation, Jagiellonian University Medical College Institute of Pediatrics, Cracow, Poland
| | - Stanisław Kwiatkowski
- Faculty of Medicine, Department of Children's Neurosurgery, Jagiellonian University Medical College Institute of Pediatrics, Cracow, Poland
| | - Marcin Majka
- Faculty of Medicine, Department of Transplantation, Jagiellonian University Medical College Institute of Pediatrics, Cracow, Poland
| |
Collapse
|
5
|
Bruschettini M, Badura A, Romantsik O. Stem cell-based interventions for the treatment of stroke in newborn infants. Cochrane Database Syst Rev 2023; 11:CD015582. [PMID: 37994736 PMCID: PMC10666199 DOI: 10.1002/14651858.cd015582.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
BACKGROUND Perinatal stroke refers to a diverse but specific group of cerebrovascular diseases that occur between 20 weeks of fetal life and 28 days of postnatal life. Acute treatment options for perinatal stroke are limited supportive care, such as controlling hypoglycemia and seizures. Stem cell-based therapies offer a potential therapeutic approach to repair, restore, or regenerate injured brain tissue. Preclinical findings have culminated in ongoing human neonatal studies. OBJECTIVES To evaluate the benefits and harms of stem cell-based interventions for the treatment of stroke in newborn infants compared to control (placebo or no treatment) or stem-cell based interventions of a different type or source. SEARCH METHODS We searched CENTRAL, PubMed, Embase, and three trials registries in February 2023. We planned to search the reference lists of included studies and relevant systematic reviews for studies not identified by the database searches. SELECTION CRITERIA We attempted to include randomized controlled trials, quasi-randomized controlled trials, and cluster trials that evaluated any of the following comparisons. • Stem cell-based interventions (any type) versus control (placebo or no treatment) • Mesenchymal stem/stromal cells (MSCs) of a specifictype (e.g. number of doses or passages) or source (e.g. autologous/allogeneic or bone marrow/cord) versus MSCs of another type or source • Stem cell-based interventions (other than MSCs) of a specific type (e.g. mononuclear cells, oligodendrocyte progenitor cells, neural stem cells, hematopoietic stem cells, or induced pluripotent stem cell-derived cells) or source (e.g. autologous/allogeneic or bone marrow/cord) versus stem cell-based interventions (other than MSCs) of another type or source • MSCs versus stem cell-based interventions other than MSCs We planned to include all types of transplantation regardless of cell source (bone marrow, cord blood, Wharton's jelly, placenta, adipose tissue, peripheral blood), type of graft (autologous or allogeneic), and dose. DATA COLLECTION AND ANALYSIS We used standard Cochrane methods. Our primary outcomes were all-cause neonatal mortality, major neurodevelopmental disability, and immune rejection or any serious adverse event. Our secondary outcomes included all-cause mortality prior to first hospital discharge, seizures, adverse effects, and death or major neurodevelopmental disability at 18 to 24 months of age. We planned to use GRADE to assess the certainty of evidence for each outcome. MAIN RESULTS We identified no completed or ongoing randomized trials that met our inclusion criteria. We excluded three studies: two were phase 1 trials, and one included newborn infants with conditions other than stroke (i.e. cerebral ischemia and anemia). Among the three excluded studies, we identified the first phase 1 trial on the use of stem cells for neonatal stroke. It reported that a single intranasal application of bone marrow-derived MSCs in term neonates with a diagnosis of perinatal arterial ischemic stroke (PAIS) was feasible and apparently not associated with severe adverse events. However, the trial included only 10 infants, and follow-up was limited to three months. AUTHORS' CONCLUSIONS No evidence is currently available to evaluate the benefits and harms of stem cell-based interventions for treatment of stroke in newborn infants. We identified no ongoing studies. Future clinical trials should focus on standardizing the timing and method of cell delivery and cell processing to optimize the therapeutic potential of stem cell-based interventions and safety profiles. Phase 1 and large animal studies might provide the groundwork for future randomized trials. Outcome measures should include all-cause mortality, major neurodevelopmental disability and immune rejection, and any other serious adverse events.
Collapse
Affiliation(s)
- Matteo Bruschettini
- Paediatrics, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Department of Research and Education, Lund University, Skåne University Hospital, Lund, Sweden
| | - Anna Badura
- Department of Neonatology, University Children's Hospital Regensburg, Hospital St Hedwig of the Order of St John, University of Regensburg, Regensburg, Germany
| | - Olga Romantsik
- Paediatrics, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
6
|
Huang S, Liu L, Huang Y, Fu C, Peng T, Yang X, Zhou H, Zhao Y, Xu Y, Zeng X, Zeng P, Tang H, He L, Xu K. Potential optimized route for mesenchymal stem cell transplantation in a rat model of cerebral palsy. Exp Cell Res 2023; 430:113734. [PMID: 37532123 DOI: 10.1016/j.yexcr.2023.113734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/04/2023] [Accepted: 07/26/2023] [Indexed: 08/04/2023]
Abstract
Cerebral palsy (CP) is a movement and posture disorder that affects over 50 million people worldwide. Human umbilical cord-derived mesenchymal stem cell (hUC-MSC) transplantation has emerged as an attractive therapeutic strategy for CP. The administration route appears to be crucial for hUC-MSC to provide adequate neuroprotection. Wistar rats were given hypoxia-ischemia to make the CP model on postnatal day 5. On postnatal day 21, DiR-labeled hUC-MSC were transplanted into the CP rats by intravenous, intrathecal, and lateral ventricle for cell tracking. Uninfused CP rats served as the negative control. The motor behavioral and pathological alteration was analyzed 11, 25, and 39 days after transplantation to assess motor function, immune inflammation, neurotrophy, and endogenous repair. In vivo imaging tracking techniques revealed that intravenous infusion resulted in fewer transplanted cells in the target brain than intrathecal and lateral ventricle infusion (p<0.05). Three different routes of hUC-MSC infusion improved the motor function of CP rats (p<0.05). At 11 days post-infusion, intrathecal infusion outperformed intravenous with a significant neurotrophic and oligodendrocyte maturation effect (p<0.05). Intrathecal infusion equaled lateral ventricle infusion after 25 days. At 39 days post-infusion, lateral ventricle infusion exceeded intravenous and intrathecal infusion with a significant immunosuppressive effect (p<0.05). Considering the improved effect and less trauma shown early in the intrathecal infusion, repeated intrathecal administration may ultimately lead to the greatest benefit.
Collapse
Affiliation(s)
- Shiya Huang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China; Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510120, China; School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Liru Liu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China; Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510120, China
| | - Yuan Huang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China; Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510120, China; School of Medicine, South China University of Technology, Guangzhou, 510655, China
| | - Chaoqiong Fu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China; Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510120, China; School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Tingting Peng
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China; Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510120, China
| | - Xubo Yang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China; Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510120, China
| | - Hongyu Zhou
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China; Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510120, China
| | - Yiting Zhao
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China; Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510120, China
| | - Yi Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China; Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510120, China
| | - Xiaoli Zeng
- Guangdong Xiangxue Stem Cell Regenerative Medicine Technology Co., Ltd, Guangzhou, 510120, China
| | - Peishan Zeng
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China; Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510120, China
| | - Hongmei Tang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China; Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510120, China
| | - Lu He
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China; Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510120, China.
| | - Kaishou Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China; Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510120, China.
| |
Collapse
|
7
|
Otsu Y, Hatakeyama M, Kanayama T, Akiyama N, Ninomiya I, Omae K, Kato T, Onodera O, Fukushima M, Shimohata T, Kanazawa M. Oxygen-Glucose Deprived Peripheral Blood Mononuclear Cells Protect Against Ischemic Stroke. Neurotherapeutics 2023; 20:1369-1387. [PMID: 37335500 PMCID: PMC10480381 DOI: 10.1007/s13311-023-01398-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2023] [Indexed: 06/21/2023] Open
Abstract
Stroke is the leading cause of severe long-term disability. Cell therapy has recently emerged as an approach to facilitate functional recovery in stroke. Although administration of peripheral blood mononuclear cells preconditioned by oxygen-glucose deprivation (OGD-PBMCs) has been shown to be a therapeutic strategy for ischemic stroke, the recovery mechanisms remain largely unknown. We hypothesised that cell-cell communications within PBMCs and between PBMCs and resident cells are necessary for a polarising protective phenotype. Here, we investigated the therapeutic mechanisms underlying the effects of OGD-PBMCs through the secretome. We compared levels of transcriptomes, cytokines, and exosomal microRNA in human PBMCs by RNA sequences, Luminex assay, flow cytometric analysis, and western blotting under normoxic and OGD conditions. We also performed microscopic analyses to assess the identification of remodelling factor-positive cells and evaluate angiogenesis, axonal outgrowth, and functional recovery by blinded examination by administration of OGD-PBMCs after ischemic stroke in Sprague-Dawley rats. We found that the therapeutic potential of OGD-PBMCs was mediated by a polarised protective state through decreased levels of exosomal miR-155-5p, and upregulation of vascular endothelial growth factor and a pluripotent stem cell marker stage-specific embryonic antigen-3 through the hypoxia-inducible factor-1α axis. After administration of OGD-PBMCs, microenvironment changes in resident microglia by the secretome promoted angiogenesis and axonal outgrowth, resulting in functional recovery after cerebral ischemia. Our findings revealed the mechanisms underlying the refinement of the neurovascular unit by secretome-mediated cell-cell communications through reduction of miR-155-5p from OGD-PBMCs, highlighting the therapeutic potential carrier of this approach against ischemic stroke.
Collapse
Affiliation(s)
- Yutaka Otsu
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-Dori, Chuoku, Niigata, 951-8585, Japan
| | - Masahiro Hatakeyama
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-Dori, Chuoku, Niigata, 951-8585, Japan
| | - Takeshi Kanayama
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-Dori, Chuoku, Niigata, 951-8585, Japan
| | - Natsuki Akiyama
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-Dori, Chuoku, Niigata, 951-8585, Japan
| | - Itaru Ninomiya
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-Dori, Chuoku, Niigata, 951-8585, Japan
| | - Kaoru Omae
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, 1-5-4 Minatojima-Minamimachi, Kobe, 650-0047, Japan
| | - Taisuke Kato
- Department of System Pathology for Neurological Disorders, Brain Science Branch, Brain Research Institute, Niigata University, 1-757 Asahimachi-Dori, Chuoku, Niigata, 951-8585, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-Dori, Chuoku, Niigata, 951-8585, Japan
| | - Masanori Fukushima
- Foundation of Learning Health Society Institute, 8F, Nagoya Mitsui Bussan Bldg. 1-16-21 Meiekiminami, Nakamura-ku, Nagoya, 450-003, Japan
| | - Takayoshi Shimohata
- Department of Neurology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Masato Kanazawa
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-Dori, Chuoku, Niigata, 951-8585, Japan.
| |
Collapse
|
8
|
Bruschettini M, Badura A, Romantsik O. Stem cell‐based interventions for the treatment of stroke in newborn infants. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2023; 2023:CD015582. [PMCID: PMC9933426 DOI: 10.1002/14651858.cd015582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
This is a protocol for a Cochrane Review (intervention). The objectives are as follows: To evaluate the benefits and harms of stem cell‐based interventions for the treatment of stroke in newborn infants compared to control (placebo or no treatment) or stem‐cell based interventions of a different type or source.
Collapse
Affiliation(s)
| | - Matteo Bruschettini
- Department of Clinical Sciences Lund, PaediatricsLund University, Skåne University HospitalLundSweden,Cochrane SwedenLund University, Skåne University HospitalLundSweden
| | | | - Olga Romantsik
- Department of Clinical Sciences Lund, PaediatricsLund University, Skåne University HospitalLundSweden
| |
Collapse
|
9
|
Okazaki K, Nakamura S, Koyano K, Konishi Y, Kondo M, Kusaka T. Neonatal asphyxia as an inflammatory disease: Reactive oxygen species and cytokines. Front Pediatr 2023; 11:1070743. [PMID: 36776908 PMCID: PMC9911547 DOI: 10.3389/fped.2023.1070743] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023] Open
Abstract
Neonatologists resuscitate asphyxiated neonates by every available means, including positive ventilation, oxygen therapy, and drugs. Asphyxiated neonates sometimes present symptoms that mimic those of inflammation, such as fever and edema. The main pathophysiology of the asphyxia is inflammation caused by hypoxic-ischemic reperfusion. At birth or in the perinatal period, neonates may suffer several, hypoxic insults, which can activate inflammatory cells and inflammatory mediator production leading to the release of larger quantities of reactive oxygen species (ROS). This in turn triggers the production of oxygen stress-induced high mobility group box-1 (HMGB-1), an endogenous damage-associated molecular patterns (DAMPs) protein bound to toll-like receptor (TLR) -4, which activates nuclear factor-kappa B (NF-κB), resulting in the production of excess inflammatory mediators. ROS and inflammatory mediators are produced not only in activated inflammatory cells but also in non-immune cells, such as endothelial cells. Hypothermia inhibits pro-inflammatory mediators. A combination therapy of hypothermia and medications, such as erythropoietin and melatonin, is attracting attention now. These medications have both anti-oxidant and anti-inflammatory effects. As the inflammatory response and oxidative stress play a critical role in the pathophysiology of neonatal asphyxia, these drugs may contribute to improving patient outcomes.
Collapse
Affiliation(s)
- Kaoru Okazaki
- Department of Neonatology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Shinji Nakamura
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kosuke Koyano
- Maternal Perinatal Center, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Yukihiko Konishi
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Masatoshi Kondo
- Department of Neonatology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Takashi Kusaka
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| |
Collapse
|
10
|
Manohar K, Mesfin FM, Liu J, Shelley WC, Brokaw JP, Markel TA. Gut-Brain cross talk: The pathogenesis of neurodevelopmental impairment in necrotizing enterocolitis. Front Pediatr 2023; 11:1104682. [PMID: 36873645 PMCID: PMC9975605 DOI: 10.3389/fped.2023.1104682] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/23/2023] [Indexed: 02/17/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating condition of multi-factorial origin that affects the intestine of premature infants and results in high morbidity and mortality. Infants that survive contend with several long-term sequelae including neurodevelopmental impairment (NDI)-which encompasses cognitive and psychosocial deficits as well as motor, vision, and hearing impairment. Alterations in the gut-brain axis (GBA) homeostasis have been implicated in the pathogenesis of NEC and the development of NDI. The crosstalk along the GBA suggests that microbial dysbiosis and subsequent bowel injury can initiate systemic inflammation which is followed by pathogenic signaling cascades with multiple pathways that ultimately lead to the brain. These signals reach the brain and activate an inflammatory cascade in the brain resulting in white matter injury, impaired myelination, delayed head growth, and eventual downstream NDI. The purpose of this review is to summarize the NDI seen in NEC, discuss what is known about the GBA, explore the relationship between the GBA and perinatal brain injury in the setting of NEC, and finally, highlight the existing research into possible therapies to help prevent these deleterious outcomes.
Collapse
Affiliation(s)
- Krishna Manohar
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - Fikir M Mesfin
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - Jianyun Liu
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - W Christopher Shelley
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - John P Brokaw
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - Troy A Markel
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States.,Riley Hospital for Children, Indiana University Health, Indianapolis, IN, United States
| |
Collapse
|
11
|
Combination Therapy of Mesenchymal Stem Cell Transplantation and Astrocyte Ablation Improve Remyelination in a Cuprizone-Induced Demyelination Mouse Model. Mol Neurobiol 2022; 59:7278-7292. [PMID: 36175823 DOI: 10.1007/s12035-022-03036-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/13/2022] [Indexed: 10/14/2022]
Abstract
Astrocytes display an active, dual, and controversial role in multiple sclerosis (MS), a chronic inflammatory demyelination disorder. However, mesenchymal stem cells (MSCs) can affect myelination in demyelinating disorders. This study aimed to investigate the effect of single and combination therapies of astrocyte ablation and MSC transplantation on remyelination in the cuprizone (CPZ) model of MS. C57BL/6 mice were fed 0.2% CPZ diet for 12 weeks. Astrocytes were ablated twice by L-a-aminoadipate (L-AAA) at the beginning of weeks 13 and 14 whereas MSCs were injected in the corpus callosum at the beginning of week 13. Motor coordination and balance were assessed through rotarod test whereas myelin content was evaluated by Luxol-fast blue (LFB) staining and transmission electron microscopy (TEM). Glial cells were assessed by immunofluorescence staining while mRNA expression was evaluated by quantitative real-time PCR. Combination treatment of ablation of astrocytes and MSC transplantation (CPZ + MSC + L-AAA) significantly decreased motor coordination deficits better than single treatments (CPZ + MSCs or CPZ + L-AAA), in comparison to CPZ mice. In addition, L-AAA and MSCs treatment significantly enhanced remyelination compared to CPZ group. Moreover, combination therapy caused a significant decrease in the number of GFAP+ and Iba-1+ cells, whereas oligodendrocytes were significantly increased in comparison to CPZ mice. Finally, MSC administration resulted in a significant upregulation of BDNF and NGF mRNA expression levels. Our data indicate that transient ablation of astrocytes along with MSCs treatment improve remyelination through enhancing oligodendrocytes and attenuating gliosis in a chronic demyelinating mouse model of MS.
Collapse
|
12
|
Neuroprotection of Bone Marrow-Derived Mesenchymal Stem Cell-Derived Extracellular Vesicle-Enclosed miR-410 Correlates with HDAC4 Knockdown in Hypoxic-Ischemic Brain Damage. Neurochem Res 2022; 47:3150-3166. [PMID: 36028735 DOI: 10.1007/s11064-022-03670-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 06/07/2022] [Accepted: 06/23/2022] [Indexed: 10/15/2022]
Abstract
Evidence exists reporting that miR-410 may rescue neurological deficits, neuronal injury, and neuronal apoptosis after experimental hypoxic ischemia. This study aimed to explore the mechanism by which miR-410 transferred by bone marrow-derived mesenchymal stem cell-derived extracellular vesicles (BMSC-EVs) may alleviate hypoxic-ischemic brain damage (HIBD) in newborn mice. BMSCs were isolated from total bone marrow cells of femur and tibia of newborn mice, and primary neurons were extracted from the cerebral cortex of newborn mice within 24 h of birth. EVs were extracted from BMSCs transfected with the mimic or inhibitor of miR-410. Primary neurons were subjected to hypoxia and treated with overexpression (oe)-HDAC4, small interfering RNA (siRNA)-β-catenin, or Wnt pathway inhibitor and/or EV (miR-410 mimic) or EV (miR-410 inhibitor). A neonatal mouse HIBD model was established and treated with EVs. When BMSC-EVs were endocytosed by primary neurons, miR-410 was upregulated, neuronal viability was elevated, and apoptosis was inhibited. miR-410 in BMSC-EVs targeted HDAC4, thus increasing neuronal viability and reducing apoptosis. Conversely, overexpression of HDAC4 activated the Wnt pathway and enhanced the nuclear translocation of β-catenin. Treatment with miR-410-containing BMSC-EVs improved learning and memory abilities of HIBD mice while attenuating apoptosis by inactivating the Wnt pathway via targeting HDAC4. Taken together, the findings suggest that miR-410 delivered by BMSC-EVs alleviates HIBD by inhibiting HDAC4-dependent Wnt pathway activation.
Collapse
|
13
|
Hypoxic Ischemic Encephalopathy (HIE) in Term and Preterm Infants. Pril (Makedon Akad Nauk Umet Odd Med Nauki) 2022; 43:77-84. [PMID: 35451288 DOI: 10.2478/prilozi-2022-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Hypoxic-ischemic syndrome (HIS) and Hypoxic-ischemic encephalopathy (HIE) are conditions that affect term and premature babies, with different pathophysiology and different brain disorders. HIE appears in 1-6 / 1000 live births and 26/1000 live births in developing countries. 15-20% die in the early neonatal period, while surviving babies have severe neurological impairment, including cerebral palsy, epilepsy, visual and hearing impairment, cognitive impairment, intellectual, behavioural, and social disorders. The hypoxic-ischemic event occurs before, during or after birth. The reasons may be related to the mother, the way of birth, the placenta, and the newborn. The criteria for diagnosis of HIE include a combination of perinatal factors, the need for resuscitation, standard neurological examinations, neurophysiological monitoring, neuroimaging methods and biochemical markers. The most effective treatment for HIE is hypothermia in combination with pharmacological therapy. HIE and HIS are problem that still persist in developing countries due to inadequate obstetric care, neonatal resuscitation, and hypothermia. Current and emerging research for HIE examines new markers for early recognition, treatment, and appropriate neuroprotection of high-risk term and premature infants.
Collapse
|
14
|
Rayasam A, Fukuzaki Y, Vexler ZS. Microglia-leucocyte axis in cerebral ischaemia and inflammation in the developing brain. Acta Physiol (Oxf) 2021; 233:e13674. [PMID: 33991400 DOI: 10.1111/apha.13674] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 05/06/2021] [Accepted: 05/08/2021] [Indexed: 12/13/2022]
Abstract
Development of the Central Nervous System (CNS) is reliant on the proper function of numerous intricately orchestrated mechanisms that mature independently, including constant communication between the CNS and the peripheral immune system. This review summarizes experimental knowledge of how cerebral ischaemia in infants and children alters physiological communication between leucocytes, brain immune cells, microglia and the neurovascular unit (NVU)-the "microglia-leucocyte axis"-and contributes to acute and long-term brain injury. We outline physiological development of CNS barriers in relation to microglial and leucocyte maturation and the plethora of mechanisms by which microglia and peripheral leucocytes communicate during postnatal period, including receptor-mediated and intracellular inflammatory signalling, lipids, soluble factors and extracellular vesicles. We focus on the "microglia-leucocyte axis" in rodent models of most common ischaemic brain diseases in the at-term infants, hypoxic-ischaemic encephalopathy (HIE) and focal arterial stroke and discuss commonalities and distinctions of immune-neurovascular mechanisms in neonatal and childhood stroke compared to stroke in adults. Given that hypoxic and ischaemic brain damage involve Toll-like receptor (TLR) activation, we discuss the modulatory role of viral and bacterial TLR2/3/4-mediated infection in HIE, perinatal and childhood stroke. Furthermore, we provide perspective of the dynamics and contribution of the axis in cerebral ischaemia depending on the CNS maturational stage at the time of insult, and modulation independently and in consort by individual axis components and in a sex dependent ways. Improved understanding on how to modify crosstalk between microglia and leucocytes will aid in developing age-appropriate therapies for infants and children who suffered cerebral ischaemia.
Collapse
Affiliation(s)
- Aditya Rayasam
- Department of Neurology University of California San Francisco San Francisco CA USA
| | - Yumi Fukuzaki
- Department of Neurology University of California San Francisco San Francisco CA USA
| | - Zinaida S. Vexler
- Department of Neurology University of California San Francisco San Francisco CA USA
| |
Collapse
|
15
|
Basham HK, Aghoghovwia BE, Papaioannou P, Seo S, Oorschot DE. Delayed Double Treatment with Adult-Sourced Adipose-Derived Mesenchymal Stem Cells Increases Striatal Medium-Spiny Neuronal Number, Decreases Striatal Microglial Number, and Has No Subventricular Proliferative Effect, after Acute Neonatal Hypoxia-Ischemia in Male Rats. Int J Mol Sci 2021; 22:ijms22157862. [PMID: 34360638 PMCID: PMC8346138 DOI: 10.3390/ijms22157862] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/05/2021] [Accepted: 07/13/2021] [Indexed: 11/17/2022] Open
Abstract
Perinatal hypoxia-ischemia (HI) is a major cause of striatal injury. Delayed post-treatment with adult-sourced bone marrow-derived mesenchymal stem cells (BMSCs) increased the absolute number of striatal medium-spiny neurons (MSNs) following perinatal HI-induced brain injury. Yet extraction of BMSCs is more invasive and difficult compared to extraction of adipose-derived mesenchymal stem cells (AD-MSCs), which are easily sourced from subcutaneous tissue. Adult-sourced AD-MSCs are also superior to BMSCs in the treatment of adult ischemic stroke. Therefore, we investigated whether delayed post-treatment with adult-sourced AD-MSCs increased the absolute number of striatal MSNs following perinatal HI-induced brain injury. This included investigation of the location of injected AD-MSCs within the brain, which were widespread in the dorsolateral subventricular zone (dlSVZ) at 1 day after their injection. Cells extracted from adult rat tissue were verified to be stem cells by their adherence to tissue culture plastic and their expression of specific ‘cluster of differentiation’ (CD) markers. They were verified to be AD-MSCs by their ability to differentiate into adipocytes and osteocytes in vitro. Postnatal day (PN) 7/8, male Sprague-Dawley rats were exposed to either HI right-sided brain injury or no HI injury. The HI rats were either untreated (HI + Diluent), single stem cell-treated (HI + MSCs×1), or double stem cell-treated (HI + MSCs×2). Control rats that were matched-for-weight and litter had no HI injury and were treated with diluent (Uninjured + Diluent). Treatment with AD-MSCs or diluent occurred either 7 days, or 7 and 9 days, after HI. There was a significant increase in the absolute number of striatal dopamine and cyclic AMP-regulated phosphoprotein (DARPP-32)-positive MSNs in the double stem cell-treated (HI + MSCs×2) group and the normal control group compared to the HI + Diluent group at PN21. We therefore investigated two potential mechanisms for this effect of double-treatment with AD-MSCs. Specifically, did AD-MSCs: (i) increase the proliferation of cells within the dlSVZ, and (ii) decrease the microglial response in the dlSVZ and striatum? It was found that a primary repair mechanism triggered by double treatment with AD-MSCs involved significantly decreased striatal inflammation. The results may lead to the development of clinically effective and less invasive stem cell therapies for neonatal HI brain injury.
Collapse
|
16
|
Pathipati P, Lecuyer M, Faustino J, Strivelli J, Phinney DG, Vexler ZS. Mesenchymal Stem Cell (MSC)-Derived Extracellular Vesicles Protect from Neonatal Stroke by Interacting with Microglial Cells. Neurotherapeutics 2021; 18:1939-1952. [PMID: 34235636 PMCID: PMC8609070 DOI: 10.1007/s13311-021-01076-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2021] [Indexed: 02/04/2023] Open
Abstract
Mesenchymal stem cell (MSC)-based therapies are beneficial in models of perinatal stroke and hypoxia-ischemia. Mounting evidence suggests that in adult injury models, including stroke, MSC-derived small extracellular vesicles (MSC-sEV) contribute to the neuroprotective and regenerative effects of MSCs. Herein, we examined if MSC-sEV protect neonatal brain from stroke and if this effect is mediated via communication with microglia. MSC-sEV derived from bone marrow MSCs were characterized by size distribution (NanoSight™) and identity (protein markers). Studies in microglial cells isolated from the injured or contralateral cortex of postnatal day 9 (P9) mice subjected to a 3-h middle cerebral artery occlusion (tMCAO) and cultured (in vitro) revealed that uptake of fluorescently labeled MSC-sEV was significantly greater by microglia from the injured cortex vs. contralateral cortex. The cell-type-specific spatiotemporal distribution of MSC-sEV was also determined in vivo after tMCAO at P9. MSC-sEV administered at reperfusion, either by intracerebroventricular (ICV) or by intranasal (IN) routes, accumulated in the hemisphere ipsilateral to the occlusion, with differing spatial distribution 2 h, 18 h, and 72 h regardless of the administration route. By 72 h, MSC-sEV in the IN group was predominantly observed in Iba1+ cells with retracted processes and in GLUT1+ blood vessels in ischemic-reperfused regions. MSC-sEV presence in Iba1+ cells was sustained. MSC-sEV administration also significantly reduced injury volume 72 h after tMCAO in part via modulatory effects on microglial cells. Together, these data establish feasibility for MSC-sEV delivery to injured neonatal brain via a clinically relevant IN route, which affords protection during sub-acute injury phase.
Collapse
Affiliation(s)
- Praneeti Pathipati
- Department of Neurology, UCSF, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
- Department of Pediatrics, UCSF, San Francisco, CA, USA
| | - Matthieu Lecuyer
- Department of Neurology, UCSF, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Joel Faustino
- Department of Neurology, UCSF, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | | | - Donald G Phinney
- Department of Molecular Medicine, Scripps Research Institute, Jupiter, FL, USA
| | - Zinaida S Vexler
- Department of Neurology, UCSF, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA.
| |
Collapse
|
17
|
Nair S, Rocha‐Ferreira E, Fleiss B, Nijboer CH, Gressens P, Mallard C, Hagberg H. Neuroprotection offered by mesenchymal stem cells in perinatal brain injury: Role of mitochondria, inflammation, and reactive oxygen species. J Neurochem 2021; 158:59-73. [PMID: 33314066 PMCID: PMC8359360 DOI: 10.1111/jnc.15267] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022]
Abstract
Preclinical studies have shown that mesenchymal stem cells have a positive effect in perinatal brain injury models. The mechanisms that cause these neurotherapeutic effects are not entirely intelligible. Mitochondrial damage, inflammation, and reactive oxygen species are considered to be critically involved in the development of injury. Mesenchymal stem cells have immunomodulatory action and exert mitoprotective effects which attenuate production of reactive oxygen species and promote restoration of tissue function and metabolism after perinatal insults. This review summarizes the present state, the underlying causes, challenges and possibilities for effective clinical translation of mesenchymal stem cell therapy.
Collapse
Affiliation(s)
- Syam Nair
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Neuroscience and PhysiologySahlgrenska Academy, University of GothenburgGothenburgSweden
- Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Eridan Rocha‐Ferreira
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Neuroscience and PhysiologySahlgrenska Academy, University of GothenburgGothenburgSweden
- Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Bobbi Fleiss
- School of Health and Biomedical SciencesRMIT UniversityBundooraVictoriaAustralia
- Université de Paris, NeuroDiderotParisFrance
| | - Cora H Nijboer
- Department for Developmental Origins of DiseaseUniversity Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht UniversityUtrechtNetherlands
| | | | - Carina Mallard
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Neuroscience and PhysiologySahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Henrik Hagberg
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| |
Collapse
|
18
|
A Brief Review on Erythropoietin and Mesenchymal Stem Cell Therapies for Paediatric Neurological Disorders. CURRENT STEM CELL REPORTS 2021. [DOI: 10.1007/s40778-021-00189-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
19
|
Therapeutic potential of stem cells for preterm infant brain damage: Can we move from the heterogeneity of preclinical and clinical studies to established therapeutics? Biochem Pharmacol 2021; 186:114461. [PMID: 33571501 DOI: 10.1016/j.bcp.2021.114461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/27/2021] [Accepted: 02/02/2021] [Indexed: 12/17/2022]
Abstract
Acquired perinatal brain injuries are a set of conditions that remains a key challenge for neonatologists and that have significant social, emotional and financial implications for our communities. In our perspective article, we will introduce perinatal brain injury focusing specifically on the events leading to brain damage in preterm born infants and outcomes for these infants. Then we will summarize and discuss the preclinical and clinical studies testing the efficacy of stem cells as neuroprotectants in the last ten years in perinatal brain injury. There are no therapies to treat brain damage in preterm born infants and a primary finding from this review is that there is a scarcity of stem cell trials focused on overcoming brain injuries in these infants. Overall, across all forms of perinatal brain injury there is a remarkable heterogeneity in previous and on-going preclinical and clinical studies in terms of the stem cell type, animal models/patient selection, route and time of administration. Despite the quality of many of the studies this variation makes it difficult to reach a valid consensus for future developments. However, it is clear that stem cells (and stem cell derived exosomes) can reduce perinatal brain injury and our field needs to work collectively to refine an effective protocol for each type of injury. The use of standardized stem cell products and testing these products across multiple models of injury will provide a stronger framework for clinical trials development.
Collapse
|
20
|
Abstract
Ischemic brain injury is a common cause of long-term neurological deficits in children as well as adults, and no efficient treatments could reverse the sequelae in clinic till now. Stem cells have the capacity of self-renewal and multilineage differentiation. The therapeutic efficacy of stem cell transplantation for ischemic brain injury have been tested for many years. The grafts could survive and mature in the ischemic brain environment. Stem cell transplantation could improve functional recovery of ischemic brain injury models in pre-clinical trials. The potential mechanisms included cell replacement, release of neurotrophic and anti-inflammatory factors, immunoregulation as well as activation of endogenous neurogenesis. Besides, many clinical trials were conducted and some of trials already had preliminary results. From the current published data, cell transplantation for clinical application is safe and feasible. No severe adverse events and tumorigenesis were reported. While the therapeutic efficacy of stem cell therapy in clinic still needs more evidences. In this review, we overviewed the studies about stem cell therapy for ischemic brain injury. Different types of stem cells used for transplantation as well as the therapeutic mechanisms were discussed in detail. The related pre-clinical and clinical trials were summarized into two separate tables. In addition, we also discussed the unsolved problems and concerns about stem cell therapy for ischemic brain injury that need to be overcome before clinic transformation.
Collapse
Affiliation(s)
- Xiao-Li Ji
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China.,Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai, China
| | - Ling Ma
- Stem Cell Research Center, Institute of Pediatrics, Children's Hospital, Fudan University, Shanghai, China
| | - Wen-Hao Zhou
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China.,Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai, China
| | - Man Xiong
- Stem Cell Research Center, Institute of Pediatrics, Children's Hospital, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Transplantation of Neural Precursors Derived from Induced Pluripotent Cells Preserve Perineuronal Nets and Stimulate Neural Plasticity in ALS Rats. Int J Mol Sci 2020; 21:ijms21249593. [PMID: 33339362 PMCID: PMC7766921 DOI: 10.3390/ijms21249593] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/05/2020] [Accepted: 12/13/2020] [Indexed: 12/11/2022] Open
Abstract
A promising therapeutic strategy for amyotrophic lateral sclerosis (ALS) treatment is stem cell therapy. Neural progenitors derived from induced pluripotent cells (NP-iPS) might rescue or replace dying motoneurons (MNs). However, the mechanisms responsible for the beneficial effect are not fully understood. The aim here was to investigate the mechanism by studying the effect of intraspinally injected NP-iPS into asymptomatic and early symptomatic superoxide dismutase (SOD)1G93A transgenic rats. Prior to transplantation, NP-iPS were characterized in vitro for their ability to differentiate into a neuronal phenotype. Motor functions were tested in all animals, and the tissue was analyzed by immunohistochemistry, qPCR, and Western blot. NP-iPS transplantation significantly preserved MNs, slowed disease progression, and extended the survival of all treated animals. The dysregulation of spinal chondroitin sulfate proteoglycans was observed in SOD1G93A rats at the terminal stage. NP-iPS application led to normalized host genes expression (versican, has-1, tenascin-R, ngf, igf-1, bdnf, bax, bcl-2, and casp-3) and the protection of perineuronal nets around the preserved MNs. In the host spinal cord, transplanted cells remained as progenitors, many in contact with MNs, but they did not differentiate. The findings suggest that NP-iPS demonstrate neuroprotective properties by regulating local gene expression and regulate plasticity by modulating the central nervous system (CNS) extracellular matrix such as perineuronal nets (PNNs).
Collapse
|
22
|
Pisani F, Fusco C, Nagarajan L, Spagnoli C. Acute symptomatic neonatal seizures, brain injury, and long-term outcome: The role of neuroprotective strategies. Expert Rev Neurother 2020; 21:189-203. [PMID: 33176104 DOI: 10.1080/14737175.2021.1848547] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Neonatal seizures are frequent but underdiagnosed manifestations of acute brain dysfunction and an important contributor to unfavorable outcomes. Etiology and severity of brain injury are the single strongest outcome determinants. AREAS COVERED The authors will discuss the prognostic role of acute symptomatic seizures versus brain injury and the main neuroprotective and neurorestorative strategies for full-term and preterm infants. EXPERT OPINION Prolonged acute symptomatic seizures likely contribute to long-term outcomes by independently adding further brain injury to initial insults. Correct timing and dosing of therapeutic interventions, depending on etiology and gestational ages, need careful evaluation. Although promising strategies are under study, the only standard of care is whole-body therapeutic hypothermia in full-term newborns with hypoxic-ischemic encephalopathy.
Collapse
Affiliation(s)
- Francesco Pisani
- Child Neuropsychiatric Unit, Medicine and Surgery Department, University of Parma , Parma, Italy
| | - Carlo Fusco
- Child Neurology Unit, Department of Paediatrics, Azienda USL-IRCCS Di Reggio Emilia , Reggio Emilia, Italy
| | - Lakshmi Nagarajan
- Department of Neurology, Perth Children's Hospital, University of Western Australia , Perth, Australia
| | - Carlotta Spagnoli
- Child Neurology Unit, Department of Paediatrics, Azienda USL-IRCCS Di Reggio Emilia , Reggio Emilia, Italy
| |
Collapse
|
23
|
Vaes JEG, van Kammen CM, Trayford C, van der Toorn A, Ruhwedel T, Benders MJNL, Dijkhuizen RM, Möbius W, van Rijt SH, Nijboer CH. Intranasal mesenchymal stem cell therapy to boost myelination after encephalopathy of prematurity. Glia 2020; 69:655-680. [PMID: 33045105 PMCID: PMC7821154 DOI: 10.1002/glia.23919] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 12/14/2022]
Abstract
Encephalopathy of prematurity (EoP) is a common cause of long-term neurodevelopmental morbidity in extreme preterm infants. Diffuse white matter injury (dWMI) is currently the most commonly observed form of EoP. Impaired maturation of oligodendrocytes (OLs) is the main underlying pathophysiological mechanism. No therapies are currently available to combat dWMI. Intranasal application of mesenchymal stem cells (MSCs) is a promising therapeutic option to boost neuroregeneration after injury. Here, we developed a double-hit dWMI mouse model and investigated the therapeutic potential of intranasal MSC therapy. Postnatal systemic inflammation and hypoxia-ischemia led to transient deficits in cortical myelination and OL maturation, functional deficits and neuroinflammation. Intranasal MSCs migrated dispersedly into the injured brain and potently improved myelination and functional outcome, dampened cerebral inflammationand rescued OL maturation after dWMI. Cocultures of MSCs with primary microglia or OLs show that MSCs secrete factors that directly promote OL maturation and dampen neuroinflammation. We show that MSCs adapt their secretome after ex vivo exposure to dWMI milieu and identified several factors including IGF1, EGF, LIF, and IL11 that potently boost OL maturation. Additionally, we showed that MSC-treated dWMI brains express different levels of these beneficial secreted factors. In conclusion, the combination of postnatal systemic inflammation and hypoxia-ischemia leads to a pattern of developmental brain abnormalities that mimics the clinical situation. Intranasal delivery of MSCs, that secrete several beneficial factors in situ, is a promising strategy to restore myelination after dWMI and subsequently improve the neurodevelopmental outcome of extreme preterm infants in the future.
Collapse
Affiliation(s)
- Josine E G Vaes
- Department for Developmental Origins of Disease, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Caren M van Kammen
- Department for Developmental Origins of Disease, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Chloe Trayford
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Annette van der Toorn
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Torben Ruhwedel
- Electron Microscopy Core Unit, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Manon J N L Benders
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Rick M Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Wiebke Möbius
- Electron Microscopy Core Unit, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Sabine H van Rijt
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Cora H Nijboer
- Department for Developmental Origins of Disease, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
24
|
Bruschettini M, Romantsik O, Moreira A, Ley D, Thébaud B. Stem cell-based interventions for the prevention of morbidity and mortality following hypoxic-ischaemic encephalopathy in newborn infants. Cochrane Database Syst Rev 2020; 8:CD013202. [PMID: 32813884 PMCID: PMC7438027 DOI: 10.1002/14651858.cd013202.pub2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hypoxic-ischaemic encephalopathy (HIE) is a leading cause of mortality and long-term neurological sequelae, affecting thousands of children worldwide. Current therapies to treat HIE are limited to cooling. Stem cell-based therapies offer a potential therapeutic approach to repair or regenerate injured brain tissue. These preclinical findings have now culminated in ongoing human neonatal trials. OBJECTIVES To determine the efficacy and safety of stem cell-based interventions for the treatment of hypoxic-ischaemic encephalopathy (HIE) in newborn infants. SEARCH METHODS We used the standard search strategy of Cochrane Neonatal to search the Cochrane Central Register of Controlled Trials (CENTRAL; 2020, Issue 5), MEDLINE via PubMed (1966 to 8 June 2020), Embase (1980 to 8 June 2020), and CINAHL (1982 to 8 June 2020). We also searched clinical trials databases, conference proceedings, and the reference lists of retrieved articles for randomised controlled trials and quasi-randomised trials. SELECTION CRITERIA Randomised controlled trials, quasi-randomised controlled trials and cluster trials comparing 1) stem cell-based interventions (any type) compared to control (placebo or no treatment); 2) use of mesenchymal stem/stromal cells (MSCs) of type (e.g. number of doses or passages) or source (e.g. autologous versus allogeneic, or bone marrow versus cord) versus MSCs of other type or source; 3) use of stem cell-based interventions other than MSCs of type (e.g. mononuclear cells, oligodendrocyte progenitor cells, neural stem cells, hematopoietic stem cells, and inducible pluripotent stem cells) or source (e.g. autologous versus allogeneic, or bone marrow versus cord) versus stem cell-based interventions other than MSCs of other type or source; or 4) MSCs versus stem cell-based interventions other than MSCs. DATA COLLECTION AND ANALYSIS For each of the included trials, two authors independently planned to extract data (e.g. number of participants, birth weight, gestational age, type and source of MSCs or other stem cell-based interventions) and assess the risk of bias (e.g. adequacy of randomisation, blinding, completeness of follow-up). The primary outcomes considered in this review are all-cause neonatal mortality, major neurodevelopmental disability, death or major neurodevelopmental disability assessed at 18 to 24 months of age. We planned to use the GRADE approach to assess the quality of evidence. MAIN RESULTS Our search strategy yielded 616 references. Two review authors independently assessed all references for inclusion. We did not find any completed studies for inclusion. Fifteen RCTs are currently registered and ongoing. We describe the three studies we excluded. AUTHORS' CONCLUSIONS There is currently no evidence from randomised trials that assesses the benefit or harms of stem cell-based interventions for the prevention of morbidity and mortality following hypoxic-ischaemic encephalopathy in newborn infants.
Collapse
Affiliation(s)
- Matteo Bruschettini
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Lund University, Skåne University Hospital, Lund, Sweden
| | - Olga Romantsik
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | - Alvaro Moreira
- Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - David Ley
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | - Bernard Thébaud
- Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Canada
- Ottawa Hospital Research Institute, Sprott Centre for Stem Cell Research, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| |
Collapse
|
25
|
Impact of a Histone Deacetylase Inhibitor-Trichostatin A on Neurogenesis after Hypoxia-Ischemia in Immature Rats. Int J Mol Sci 2020; 21:ijms21113808. [PMID: 32471267 PMCID: PMC7312253 DOI: 10.3390/ijms21113808] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/14/2020] [Accepted: 05/26/2020] [Indexed: 12/22/2022] Open
Abstract
Hypoxia-ischemia (HI) in the neonatal brain frequently results in neurologic impairments, including cognitive disability. Unfortunately, there are currently no known treatment options to minimize ischemia-induced neural damage. We previously showed the neuroprotective/neurogenic potential of a histone deacetylase inhibitor (HDACi), sodium butyrate (SB), in a neonatal HI rat pup model. The aim of the present study was to examine the capacity of another HDACi—Trichostatin A (TSA)—to stimulate neurogenesis in the subgranular zone of the hippocampus. We also assessed some of the cellular/molecular processes that could be involved in the action of TSA, including the expression of neurotrophic factors (glial cell line-derived neurotrophic factor (GDNF), nerve growth factor (NGF), and brain-derived neurotrophic factor (BDNF)) as well as the TrkB receptor and its downstream signalling substrate— cAMP response element-binding protein (CREB). Seven-day-old rat pups were subjected to unilateral carotid artery ligation followed by hypoxia for 1 h. TSA was administered directly after the insult (0.2 mg/kg body weight). The study demonstrated that treatment with TSA restored the reduced by hypoxia-ischemia number of immature neurons (neuroblasts, BrdU/DCX-positive) as well as the number of oligodendrocyte progenitors (BrdU/NG2+) in the dentate gyrus of the ipsilateral damaged hemisphere. However, new generated cells did not develop the more mature phenotypes. Moreover, the administration of TSA stimulated the expression of BDNF and increased the activation of the TrkB receptor. These results suggest that BDNF-TrkB signalling pathways may contribute to the effects of TSA after neonatal hypoxic-ischemic injury.
Collapse
|
26
|
Liau LL, Al-Masawa ME, Koh B, Looi QH, Foo JB, Lee SH, Cheah FC, Law JX. The Potential of Mesenchymal Stromal Cell as Therapy in Neonatal Diseases. Front Pediatr 2020; 8:591693. [PMID: 33251167 PMCID: PMC7672022 DOI: 10.3389/fped.2020.591693] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/05/2020] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) can be derived from various tissue sources, such as the bone marrow (BMSCs), adipose tissue (ADSCs), umbilical cord (UC-MSCs) and umbilical cord blood (UCB-MSCs). Clinical trials have been conducted to investigate the potential of MSCs in ameliorating neonatal diseases, including bronchopulmonary dysplasia (BPD), intraventricular hemorrhage (IVH) and necrotizing enterocolitis (NEC). In preclinical studies, MSC therapy has been tested for the treatment of various neonatal diseases affecting the heart, eye, gut, and brain as well as sepsis. Up to date, the number of clinical trials using MSCs to treat neonatal diseases is still limited. The data reported thus far positioned MSC therapy as safe with positive outcomes. However, most of these trials are still preliminary and generally smaller in scale. Larger trials with more appropriate controls and a longer follow-up period need to be conducted to prove the safety and efficacy of the therapy more conclusively. This review discusses the current application of MSCs in treating neonatal diseases, its mechanism of action and future direction of this novel therapy, including the potential of using MSC-derived extracellular vesicles instead of the cells to treat various clinical conditions in the newborn.
Collapse
Affiliation(s)
- Ling Ling Liau
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Maimonah Eissa Al-Masawa
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Benson Koh
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Qi Hao Looi
- Future Cytohealth Sdn Bhd, Bandar Seri Petaling, Kuala Lumpur, Malaysia
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Sau Har Lee
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Fook Choe Cheah
- Department of Paediatrics, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Jia Xian Law
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
27
|
Abstract
Advances in neonatology have led to unprecedented improvements in neonatal survival such that those born as early as 22 weeks of gestation now have some chance of survival, and over 70% of those born at 24 weeks of gestation survive. Up to 50% of infants born extremely preterm develop poor outcomes involving long-term neurodevelopmental impairments affecting cognition and learning, or motor problems such as cerebral palsy. Poor outcomes arise because the preterm brain is vulnerable both to direct injury (by events such as intracerebral hemorrhage, infection, and/or hypoxia), or indirect injury due to disruption of normal development. This neonatal brain injury and/or dysmaturation is called "encephalopathy of prematurity". Current and future strategies to improve outcomes in this population include prevention of preterm birth, and pre-, peri-, and postnatal approaches to protect the developing brain. This review will describe mechanisms of preterm brain injury, and current and upcoming therapies in the antepartum and postnatal period to improve preterm encephalopathy.
Collapse
Affiliation(s)
- Pratik Parikh
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, WA.
| | - Sandra E Juul
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, WA.
| |
Collapse
|
28
|
Abstract
Perinatal brain injury is a major cause of neurological disability in both premature and term infants. In this review, we summarize the evidence behind some established neuroprotective practices such as administration of antenatal steroids, intrapartum magnesium for preterm delivery, and therapeutic hypothermia. In addition, we examine emerging practices such as delayed cord clamping, postnatal magnesium administration, recombinant erythropoietin, and non-steroidal anti-inflammatory agents and finally inform the reader about novel interventions, some of which are currently in trials, such as xenon, melatonin, topiramate, allopurinol, creatine, and autologous cord cell therapy.
Collapse
Affiliation(s)
- Samata Singhi
- Department of Neurology, Kennedy Krieger Institute, Baltimore, Maryland, 21205, USA
- Department of Pediatric Neurology, Johns Hopkins Medicine, Baltimore, MD, 21287, USA
| | - Michael Johnston
- Department of Neurology, Kennedy Krieger Institute, Baltimore, Maryland, 21205, USA
| |
Collapse
|
29
|
Kim YE, Sung SI, Chang YS, Ahn SY, Sung DK, Park WS. Thrombin Preconditioning Enhances Therapeutic Efficacy of Human Wharton's Jelly-Derived Mesenchymal Stem Cells in Severe Neonatal Hypoxic Ischemic Encephalopathy. Int J Mol Sci 2019; 20:E2477. [PMID: 31137455 PMCID: PMC6566845 DOI: 10.3390/ijms20102477] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 05/10/2019] [Accepted: 05/17/2019] [Indexed: 02/08/2023] Open
Abstract
We investigated whether thrombin preconditioning of human Wharton's jelly-derived mesenchymal stem cells (MSCs) improves paracrine potency and thus the therapeutic efficacy of naïve MSCs against severe hypoxic ischemic encephalopathy (HIE). Thrombin preconditioning significantly enhances the neuroprotective anti-oxidative, anti-apoptotic, and anti-cytotoxic effects of naïve MSCs against oxygen-glucose deprivation (OGD) of cortical neurons in vitro. Severe HIE was induced in vivo using unilateral carotid artery ligation and hypoxia for 2 h and confirmed using brain magnetic resonance imaging (MRI) involving >40% of ipsilateral hemisphere at postnatal day (P) 7 in newborn rats. Delayed intraventricular transplantation of 1 × 105 thrombin preconditioned but not naïve MSCs at 24 h after hypothermia significantly enhanced observed anti-inflammatory, anti-astroglial, and anti-apoptotic effects and the ensuing brain infarction; behavioral tests, such as cylinder rearing and negative geotaxis tests, were conducted at P42. In summary, thrombin preconditioning of human Wharton's jelly-derived MSCs significantly boosted the neuroprotective effects of naïve MSCs against OGD in vitro by enhancing their anti-oxidative, anti-apoptotic, and anti-cytotoxic effects, and significantly attenuated the severe HIE-induced brain infarction and improved behavioral function tests in vivo by maximizing their paracrine anti-inflammatory, anti-astroglial, and anti-apoptotic effects.
Collapse
Affiliation(s)
- Young Eun Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.
| | - Se In Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea.
| | - So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.
| | - Dong Kyung Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Korea.
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Korea.
| |
Collapse
|
30
|
Xue H, Xu Y, Wang S, Wu ZY, Li XY, Zhang YH, Niu JY, Gao QS, Zhao P. Sevoflurane post-conditioning alleviates neonatal rat hypoxic-ischemic cerebral injury via Ezh2-regulated autophagy. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:1691-1706. [PMID: 31190748 PMCID: PMC6528650 DOI: 10.2147/dddt.s197325] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/04/2019] [Indexed: 12/12/2022]
Abstract
Background: When neonatal rats suffer hypoxic-ischemic brain injury (HIBI), autophagy is over-activated in the hippocampus, and inhibition of autophagy provides neuroprotection. The aim of this study was to investigate the possible roles of autophagy and Ezh2-regulated Pten/Akt/mTOR pathway in sevoflurane post-conditioning (SPC)-mediated neuroprotection against HIBI in neonatal rats. Methods: Seven-day-old Sprague–Dawley rats underwent left common artery ligation followed by 2 h hypoxia as described in the Rice–Vannucci model. The roles of autophagy and the Ezh2-regulated Pten/Akt/mTOR signaling pathway in the neuroprotection conferred by SPC were examined by left-side intracerebroventricular injection with the autophagy activator rapamycin and the Ezh2 inhibitor GSK126. Results: SPC was neuroprotective against HIBI through the inhibition of over-activated autophagy in the hippocampus as characterized by the rapamycin-induced reversal of neuronal density, neuronal morphology, cerebral morphology, and the expression of the autophagy markers, LC3B-II and Beclin1. SPC significantly increased the expression of Ezh2, H3K27me3, pAkt, and mTOR and decreased the expression of Pten induced by HI. The Ezh2 inhibitor, GSK126, significantly reversed the SPC-induced changes in expression of H3K27me3, Pten, pAkt, mTOR, LC3B-II, and Beclin1. Ezh2 inhibition also reversed SPC-mediated attenuation of neuronal loss and behavioral improvement in the Morris water maze. Conclusion: These results indicate that SPC inhibits excessive autophagy via the regulation of Pten/Akt/mTOR signaling by Ezh2 to confer neuroprotection against HIBI in neonatal rats.
Collapse
Affiliation(s)
- Hang Xue
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| | - Ying Xu
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| | - Shuo Wang
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| | - Zi-Yi Wu
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| | - Xing-Yue Li
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| | - Ya-Han Zhang
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| | - Jia-Yuan Niu
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| | - Qiu-Shi Gao
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, People's Republic of China
| |
Collapse
|
31
|
Vaes JEG, Vink MA, de Theije CGM, Hoebeek FE, Benders MJNL, Nijboer CHA. The Potential of Stem Cell Therapy to Repair White Matter Injury in Preterm Infants: Lessons Learned From Experimental Models. Front Physiol 2019; 10:540. [PMID: 31143126 PMCID: PMC6521595 DOI: 10.3389/fphys.2019.00540] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022] Open
Abstract
Diffuse white matter injury (dWMI) is a major cause of morbidity in the extremely preterm born infant leading to life-long neurological impairments, including deficits in cognitive, motor, sensory, psychological, and behavioral functioning. At present, no treatment options are clinically available to combat dWMI and therefore exploration of novel strategies is urgently needed. In recent years, the pathophysiology underlying dWMI has slowly started to be unraveled, pointing towards the disturbed maturation of oligodendrocytes (OLs) as a key mechanism. Immature OL precursor cells in the developing brain are believed to be highly sensitive to perinatal inflammation and cerebral oxygen fluctuations, leading to impaired OL differentiation and eventually myelination failure. OL lineage development under normal and pathological circumstances and the process of (re)myelination have been studied extensively over the years, often in the context of other adult and pediatric white matter pathologies such as stroke and multiple sclerosis (MS). Various studies have proposed stem cell-based therapeutic strategies to boost white matter regeneration as a potential strategy against a wide range of neurological diseases. In this review we will discuss experimental studies focusing on mesenchymal stem cell (MSC) therapy to reduce white matter injury (WMI) in multiple adult and neonatal neurological diseases. What lessons have been learned from these previous studies and how can we translate this knowledge to application of MSCs for the injured white matter in the preterm infant? A perspective on the current state of stem cell therapy will be given and we will discuss different important considerations of MSCs including cellular sources, timing of treatment and administration routes. Furthermore, we reflect on optimization strategies that could potentially reinforce stem cell therapy, including preconditioning and genetic engineering of stem cells or using cell-free stem cell products, to optimize cell-based strategy for vulnerable preterm infants in the near future.
Collapse
Affiliation(s)
- Josine E G Vaes
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Marit A Vink
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Caroline G M de Theije
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Freek E Hoebeek
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Manon J N L Benders
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Cora H A Nijboer
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
32
|
Safety and Efficacy of Intraventricular Delivery of Bone Marrow-Derived Mesenchymal Stem Cells in Hemorrhagic Stroke Model. Sci Rep 2019; 9:5674. [PMID: 30952961 PMCID: PMC6450980 DOI: 10.1038/s41598-019-42182-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/21/2019] [Indexed: 01/01/2023] Open
Abstract
External ventricular drain (EVD) is used clinically to relieve intracranial pressure and occasionally to deliver medications following intracerebral hemorrhage (ICH). Mesenchymal stem cell (MSC) therapy has been shown to be neuroprotective and can induce neuroregeneration in stroke models. We evaluated the safety and efficacy of delivering MSCs intraventricularly in a rat hemorrhagic stroke model. Using autologous blood, hemorrhagic stroke was induced at specific coordinates in the right basal ganglia. After 30 minutes, rats were treated with either bone marrow-derived MSCs or a phosphate-buffered saline placebo via direct intraventricular infusion. Three dosages (2 × 105/kg, 5 × 105/kg, and 1 × 106/kg) of MSCs were administered. Forelimb use asymmetry test was employed to evaluate functional improvement after cell therapy. At the end of the experiment, peripheral blood samples and organs were harvested; biochemistry, cytokine, and growth factor analysis and histology evaluations were performed to explore cell toxicity and cell fate, and the effects of MSC therapy on injury volume, anti-inflammation, and neurogenesis. Intraventricular administration of MSCs in ICH rat model showed improved behavior and alleviated brain damage. Additionally, treated ICH rats showed significantly reduced expression of IL-1α, IL-6, and IFN-γ. No obvious cell toxicity was noticed through blood chemistry and histology evaluation. None of the infused MSCs were detected at the end of the experiment. EVD is safe and effective to use as a method of delivering MSCs to treat ICH. Intraventricularly delivered MSCs have anti-inflammatory properties and a capacity to induce neurogenesis and improve function following ICH injury.
Collapse
|
33
|
Sherman LS, Romagano MP, Williams SF, Rameshwar P. Mesenchymal stem cell therapies in brain disease. Semin Cell Dev Biol 2019; 95:111-119. [PMID: 30922957 DOI: 10.1016/j.semcdb.2019.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/08/2019] [Accepted: 03/19/2019] [Indexed: 12/19/2022]
Abstract
As treatments for diseases throughout the body progress, treatment for many brain diseases has been at a standstill due to difficulties in drug delivery. While new drugs are being discovered in vitro, these therapies are often hindered by inefficient tissue distribution and, more commonly, an inability to cross the blood brain barrier. Mesenchymal stem cells are thus being investigated as a delivery tool to directly target therapies to the brain to treat wide array of brain diseases. This review discusses the use of mesenchymal stem cells in hypoxic disease (hypoxic ischemic encephalopathy), an inflammatory neurodegenerative disease (multiple sclerosis), and a malignant condition (glioma).
Collapse
Affiliation(s)
- Lauren S Sherman
- Division of Hematology/Oncology, Department of Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA; School of Graduate Studies, Biomedical Sciences Programs - Newark, Rutgers University, Newark, NJ, USA
| | - Matthew P Romagano
- Department of Obstetrics, Gynecology and Women's Health, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Shauna F Williams
- Department of Obstetrics, Gynecology and Women's Health, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Pranela Rameshwar
- Division of Hematology/Oncology, Department of Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA.
| |
Collapse
|
34
|
van Bel F, Vaes J, Groenendaal F. Prevention, Reduction and Repair of Brain Injury of the Preterm Infant. Front Physiol 2019; 10:181. [PMID: 30949060 PMCID: PMC6435588 DOI: 10.3389/fphys.2019.00181] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 02/14/2019] [Indexed: 12/15/2022] Open
Affiliation(s)
- Frank van Bel
- Department of Neonatology, Wilhelmina Children’s Hospital and Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Josine Vaes
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Floris Groenendaal
- Department of Neonatology, Wilhelmina Children’s Hospital and Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
35
|
Farina M, Alexander JF, Thekkedath U, Ferrari M, Grattoni A. Cell encapsulation: Overcoming barriers in cell transplantation in diabetes and beyond. Adv Drug Deliv Rev 2019; 139:92-115. [PMID: 29719210 DOI: 10.1016/j.addr.2018.04.018] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/19/2018] [Accepted: 04/25/2018] [Indexed: 02/07/2023]
Abstract
Cell-based therapy is emerging as a promising strategy for treating a wide range of human diseases, such as diabetes, blood disorders, acute liver failure, spinal cord injury, and several types of cancer. Pancreatic islets, blood cells, hepatocytes, and stem cells are among the many cell types currently used for this strategy. The encapsulation of these "therapeutic" cells is under intense investigation to not only prevent immune rejection but also provide a controlled and supportive environment so they can function effectively. Some of the advanced encapsulation systems provide active agents to the cells and enable a complete retrieval of the graft in the case of an adverse body reaction. Here, we review various encapsulation strategies developed in academic and industrial settings, including the state-of-the-art technologies in advanced preclinical phases as well as those undergoing clinical trials, and assess their advantages and challenges. We also emphasize the importance of stimulus-responsive encapsulated cell systems that provide a "smart and live" therapeutic delivery to overcome barriers in cell transplantation as well as their use in patients.
Collapse
|
36
|
Bruschettini M, Romantsik O, Moreira A, Ley D, Thébaud B. Stem cell-based interventions for the prevention of morbidity and mortality following hypoxic-ischaemic encephalopathy in newborn infants. Hippokratia 2018. [DOI: 10.1002/14651858.cd013202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Matteo Bruschettini
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
- Skåne University Hospital; Cochrane Sweden; Wigerthuset, Remissgatan 4, first floor room 11-221 Lund Sweden 22185
| | - Olga Romantsik
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
| | - Alvaro Moreira
- University of Texas Health Science Center at San Antonio; Pediatrics, Division of Neonatology; San Antonio Texas USA
| | - David Ley
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
| | - Bernard Thébaud
- Children's Hospital of Eastern Ontario; Department of Pediatrics; Ottawa ON Canada
- Ottawa Hospital Research Institute, Sprott Center for Stem Cell Research; Ottawa Canada
- University of Ottawa; Department of Cellular and Molecular Medicine; Ottawa Canada
| |
Collapse
|
37
|
He M, Shi X, Yang M, Yang T, Li T, Chen J. Mesenchymal stem cells-derived IL-6 activates AMPK/mTOR signaling to inhibit the proliferation of reactive astrocytes induced by hypoxic-ischemic brain damage. Exp Neurol 2018; 311:15-32. [PMID: 30213506 DOI: 10.1016/j.expneurol.2018.09.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/10/2018] [Accepted: 09/07/2018] [Indexed: 01/08/2023]
Abstract
Mesenchymal stem cells (MSCs) treatment is an effective strategy for the functional repair of central nervous system (CNS) insults through the production of bioactive molecules. We have previously demonstrated that the interleukin-6 (IL-6) secreted by MSCs plays an anti-apoptotic role in injured astrocytes and partly promotes functional recovery in neonatal rats with hypoxic-ischemic brain damage (HIBD). However, the mechanisms of IL-6 underlying the proliferation of injured astrocytes have not been fully elucidated. In this study, we investigated the therapeutic effects of MSCs on astrocyte proliferation in neonatal rats subjected to HIBD. A HIBD model was established in Sprague Dawley (SD) rats, and MSCs were administered by intracerebroventricular injection 5 days after HIBD. Rat primary astrocytes were cultured, subjected to oxygen glucose deprivation (OGD) injury and then immediately co-cultured with MSCs in vitro. Immunofluorescence staining, Cell Counting Kit (CCK)-8, flow cytometry, Ca2+ imaging, enzyme-linked immunosorbent assay (ELISA), western blotting, and co-immunoprecipitation (Co-IP) were performed. We found that MSCs transplantation not only promoted the recovery of learning and memory function in HIBD rats but also significantly reduced the number of Ki67+/glial fibrillary acidic protein (GFAP)+ cells in the hippocampi 7-14 days after HIBD. In addition to increasing IL-6 expression in both the hippocampi of HIBD rats and astrocyte culture medium, MSCs treatment in vitro significantly increased the expression levels of glycoprotein (gp) 130 and phosphorylated AMP-activated protein kinase α (p-AMPKα) and decreased the expression levels of p-mammalian target of rapamycin (mTOR) and its downstream targets. Furthermore, MSCs treatment induced a protein-protein interaction between gp130 and p-AMPKα. Suppression of IL-6 expression in MSCs reversed the above regulatory functions of MSCs in hippocampal astrocytes. The utilization of rapamycin further confirmed that mTOR participated in the proliferation of reactive astrocytes. These findings suggest that endogenous IL-6 produced by MSCs in the HIBD microenvironment provides therapeutic advantages by activating AMPK/mTOR signaling, thus reducing the proliferation of reactive astrocytes.
Collapse
Affiliation(s)
- Mulan He
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China; Chongqing Stem Cell Therapy Engineering Technical Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Xia Shi
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China; Chongqing Stem Cell Therapy Engineering Technical Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Miao Yang
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China; Chongqing Stem Cell Therapy Engineering Technical Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Ting Yang
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Tingyu Li
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Jie Chen
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China; Chongqing Stem Cell Therapy Engineering Technical Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China.
| |
Collapse
|
38
|
Shin JE, Jung K, Kim M, Hwang K, Lee H, Kim IS, Lee BH, Lee IS, Park KI. Brain and spinal cord injury repair by implantation of human neural progenitor cells seeded onto polymer scaffolds. Exp Mol Med 2018; 50:1-18. [PMID: 29674624 PMCID: PMC5938022 DOI: 10.1038/s12276-018-0054-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 12/18/2017] [Accepted: 12/21/2017] [Indexed: 12/22/2022] Open
Abstract
Hypoxic-ischemic (HI) brain injury and spinal cord injury (SCI) lead to extensive tissue loss and axonal degeneration. The combined application of the polymer scaffold and neural progenitor cells (NPCs) has been reported to enhance neural repair, protection and regeneration through multiple modes of action following neural injury. This study investigated the reparative ability and therapeutic potentials of biological bridges composed of human fetal brain-derived NPCs seeded upon poly(glycolic acid)-based scaffold implanted into the infarction cavity of a neonatal HI brain injury or the hemisection cavity in an adult SCI. Implantation of human NPC (hNPC)–scaffold complex reduced the lesion volume, induced survival, engraftment, and differentiation of grafted cells, increased neovascularization, inhibited glial scar formation, altered the microglial/macrophage response, promoted neurite outgrowth and axonal extension within the lesion site, and facilitated the connection of damaged neural circuits. Tract tracing demonstrated that hNPC–scaffold grafts appear to reform the connections between neurons and their targets in both cerebral hemispheres in HI brain injury and protect some injured corticospinal fibers in SCI. Finally, the hNPC–scaffold complex grafts significantly improved motosensory function and attenuated neuropathic pain over that of the controls. These findings suggest that, with further investigation, this optimized multidisciplinary approach of combining hNPCs with biomaterial scaffolds provides a more versatile treatment for brain injury and SCI. Biodegradable scaffolds seeded with human fetal brain cells can help repair neurological injuries in rodents. A team led by Kook In Park and Il-Shin Lee from the Yonsei University College of Medicine in Seoul, South Korea, created a mesh of plastic fibers that they bathed in neural progenitor cells. Over the course of several days, these cells differentiated into different types of brain cells, including neurons and glia. The researchers implanted these cell-scaffold complexes into the sites of injury in two rodent models: newborn mice with oxygen deprivation to the brain, and adult rats with severed spinal cords. In both cases, the treatment helped the injured tissues heal and improved the neurological or motor function of the animals. The authors suggest these tissue-engineered structures could also help people with brain or spine injuries.
Collapse
Affiliation(s)
- Jeong Eun Shin
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Kwangsoo Jung
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Miri Kim
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Kyujin Hwang
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Haejin Lee
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Il-Sun Kim
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Bae Hwan Lee
- Department of Physiology, Brain Research Institute, Epilepsy Research Institute, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Il-Shin Lee
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.
| | - Kook In Park
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, 03722, Korea. .,Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Korea. .,Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.
| |
Collapse
|
39
|
The Potentials and Caveats of Mesenchymal Stromal Cell-Based Therapies in the Preterm Infant. Stem Cells Int 2018; 2018:9652897. [PMID: 29765429 PMCID: PMC5911321 DOI: 10.1155/2018/9652897] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 03/04/2018] [Indexed: 02/06/2023] Open
Abstract
Preponderance of proinflammatory signals is a characteristic feature of all acute and resulting long-term morbidities of the preterm infant. The proinflammatory actions are best characterized for bronchopulmonary dysplasia (BPD) which is the chronic lung disease of the preterm infant with lifelong restrictions of pulmonary function and severe consequences for psychomotor development and quality of life. Besides BPD, the immature brain, eye, and gut are also exposed to inflammatory injuries provoked by infection, mechanical ventilation, and oxygen toxicity. Despite the tremendous progress in the understanding of disease pathologies, therapeutic interventions with proven efficiency remain restricted to a few drug therapies with restricted therapeutic benefit, partially considerable side effects, and missing option of applicability to the inflamed brain. The therapeutic potential of mesenchymal stromal cells (MSCs)—also known as mesenchymal stem cells—has attracted much attention during the recent years due to their anti-inflammatory activities and their secretion of growth and development-promoting factors. Based on a molecular understanding, this review summarizes the positive actions of exogenous umbilical cord-derived MSCs on the immature lung and brain and the therapeutic potential of reprogramming resident MSCs. The pathomechanistic understanding of MSC actions from the animal model is complemented by the promising results from the first phase I clinical trials testing allogenic MSC transplantation from umbilical cord blood. Despite all the enthusiasm towards this new therapeutic option, the caveats and outstanding issues have to be critically evaluated before a broad introduction of MSC-based therapies.
Collapse
|
40
|
Bennet L, Dhillon S, Lear CA, van den Heuij L, King V, Dean JM, Wassink G, Davidson JO, Gunn AJ. Chronic inflammation and impaired development of the preterm brain. J Reprod Immunol 2018; 125:45-55. [DOI: 10.1016/j.jri.2017.11.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 11/13/2017] [Accepted: 11/24/2017] [Indexed: 12/17/2022]
|
41
|
Nijboer CH, Kooijman E, van Velthoven CT, van Tilborg E, Tiebosch IA, Eijkelkamp N, Dijkhuizen RM, Kesecioglu J, Heijnen CJ. Intranasal Stem Cell Treatment as a Novel Therapy for Subarachnoid Hemorrhage. Stem Cells Dev 2018; 27:313-325. [PMID: 29310519 DOI: 10.1089/scd.2017.0148] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) represents a major health problem in Western society due to high mortality and morbidity, and the relative young age of patients. Currently, efficacious therapeutic options are very limited. Mesenchymal stem cell (MSC) administration has been shown to improve functional outcome and lesion size in experimental models of stroke and neonatal hypoxic-ischemic brain injury. Here, we studied the therapeutic potential of intranasally administered bone marrow-derived MSCs relatively late postinsult using a rat endovascular puncture model for SAH. Six days after induction of SAH, rats were treated with MSCs or vehicle through nasal administration. Intranasal MSC treatment significantly improved sensorimotor and mechanosensory function at 21 days after SAH. Gray and white matter loss was significantly reduced by MSC treatment and the number of NeuN+ neurons around the lesion increased due to MSC treatment. Moreover, intranasal MSC administration led to a sharp decrease in SAH-induced activation of astrocytes and microglia/macrophages in the lesioned hemisphere, especially of M2-like (CD206+) microglia/macrophages. Interestingly, MSC administration also decreased SAH-induced depression-like behavior in association with a restoration of tyrosine hydroxylase expression in the substantia nigra and striatum. We show here for the first time that intranasal MSC administration reverses the devastating consequences of SAH, including regeneration of the cerebral lesion, functional recovery, and treatment of comorbid depression-like behavior.
Collapse
Affiliation(s)
- Cora H Nijboer
- 1 Laboratory of Neuroimmunology and Developmental Origins of Disease, Division Woman and Baby, University Medical Center Utrecht , Utrecht, the Netherlands
| | - Elke Kooijman
- 1 Laboratory of Neuroimmunology and Developmental Origins of Disease, Division Woman and Baby, University Medical Center Utrecht , Utrecht, the Netherlands .,2 Department of Intensive Care Medicine, University Medical Center Utrecht , Utrecht, the Netherlands
| | - Cindy T van Velthoven
- 1 Laboratory of Neuroimmunology and Developmental Origins of Disease, Division Woman and Baby, University Medical Center Utrecht , Utrecht, the Netherlands
| | - Erik van Tilborg
- 1 Laboratory of Neuroimmunology and Developmental Origins of Disease, Division Woman and Baby, University Medical Center Utrecht , Utrecht, the Netherlands
| | - Ivo A Tiebosch
- 3 Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht , Utrecht, the Netherlands
| | - Niels Eijkelkamp
- 1 Laboratory of Neuroimmunology and Developmental Origins of Disease, Division Woman and Baby, University Medical Center Utrecht , Utrecht, the Netherlands
| | - Rick M Dijkhuizen
- 3 Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht , Utrecht, the Netherlands
| | - Jozef Kesecioglu
- 2 Department of Intensive Care Medicine, University Medical Center Utrecht , Utrecht, the Netherlands
| | - Cobi J Heijnen
- 4 Laboratory of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center , Houston, Texas
| |
Collapse
|
42
|
Promoting neuroregeneration after perinatal arterial ischemic stroke: neurotrophic factors and mesenchymal stem cells. Pediatr Res 2018; 83:372-384. [PMID: 28949952 DOI: 10.1038/pr.2017.243] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/19/2017] [Indexed: 01/02/2023]
Abstract
Newborns suffering from perinatal arterial ischemic stroke (PAIS) are at risk of neurodevelopmental problems. Current treatment options for PAIS are limited and mainly focus on supportive care, as presentation of PAIS is beyond the time window of current treatment strategies. Therefore, recent focus has shifted to interventions that stimulate regeneration of damaged brain tissue. From animal models, it is known that the brain increases its neurogenic capability after ischemic injury, by promoting neural cell proliferation and differentiation. However, neurogenesis is not maintained at the long term, which consequently impedes full repair leading to adverse consequences later in life. Boosting neuroregeneration of the newborn brain using treatment with neurotrophic factors and/or mesenchymal stem cells (MSCs) may be promising novel therapeutic strategies to improve neurological prospects and quality of life of infants with PAIS. This review focuses on effectiveness of neurotrophic growth factors, including erythropoietin, brain-derived neurotrophic factor, vascular endothelial growth factor, glial-derived neurotrophic factor, and MSC therapy, in both experimental neonatal stroke studies and first clinical trials for neonatal ischemic brain injury.
Collapse
|
43
|
Strategies to enhance paracrine potency of transplanted mesenchymal stem cells in intractable neonatal disorders. Pediatr Res 2018; 83:214-222. [PMID: 28972960 DOI: 10.1038/pr.2017.249] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/21/2017] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cell (MSC) transplantation represents the next breakthrough in the treatment of currently intractable and devastating neonatal disorders with complex multifactorial etiologies, including bronchopulmonary dysplasia, hypoxic ischemic encephalopathy, and intraventricular hemorrhage. Absent engraftment and direct differentiation of transplanted MSCs, and the "hit-and-run" therapeutic effects of these MSCs suggest that their pleiotropic protection might be attributable to paracrine activity via the secretion of various biologic factors rather than to regenerative activity. The transplanted MSCs, therefore, exert their therapeutic effects not by acting as "stem cells," but rather by acting as "paracrine factors factory." The MSCs sense the microenvironment of the injury site and secrete various paracrine factors that serve several reparative functions, including antiapoptotic, anti-inflammatory, antioxidative, antifibrotic, and/or antibacterial effects in response to environmental cues to enhance regeneration of the damaged tissue. Therefore, the therapeutic efficacy of MSCs might be dependent on their paracrine potency. In this review, we focus on recent investigations that elucidate the specifically regulated paracrine mechanisms of MSCs by injury type and discuss potential strategies to enhance paracrine potency, and thus therapeutic efficacy, of transplanted MSCs, including determining the appropriate source and preconditioning strategy for MSCs and the route and timing of their administration.
Collapse
|
44
|
Parikh P, Juul SE. Neuroprotective Strategies in Neonatal Brain Injury. J Pediatr 2018; 192:22-32. [PMID: 29031859 DOI: 10.1016/j.jpeds.2017.08.031] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 06/26/2017] [Accepted: 08/15/2017] [Indexed: 01/11/2023]
Affiliation(s)
- Pratik Parikh
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, WA
| | - Sandra E Juul
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, WA.
| |
Collapse
|
45
|
Wagenaar N, Nijboer CH, van Bel F. Repair of neonatal brain injury: bringing stem cell-based therapy into clinical practice. Dev Med Child Neurol 2017; 59:997-1003. [PMID: 28786482 DOI: 10.1111/dmcn.13528] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/07/2017] [Indexed: 12/15/2022]
Abstract
Hypoxic-ischaemic brain injury is one of most important causes of neonatal mortality and long-term neurological morbidity in infants born at term. At present, only hypothermia in infants with perinatal hypoxic-ischaemic encephalopathy has shown benefit as a neuroprotective strategy. Otherwise, current treatment options for neonatal brain injury mainly focus on controlling (associated) symptoms. Regeneration of the injured neonatal brain with stem cell-based therapies is emerging and experimental results are promising. At present, increasing efforts are made to bring stem cell-based therapies to the clinic. Among all progenitor cell types, mesenchymal stromal (stem) cells seem to be most promising for human use given their neuroregenerative properties and favourable safety profile. This review summarizes the actual state, potential hurdles and possibilities of stem cell-based therapy for neonatal brain injury in the clinical setting. An early version of this paper was presented at the Groningen Early Intervention Meeting which was held in April 2016.
Collapse
Affiliation(s)
- Nienke Wagenaar
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Cora H Nijboer
- Laboratory of NeuroImmunology and Developmental Origins of Disease, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Frank van Bel
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| |
Collapse
|
46
|
Dai Y, Li W, Zhong M, Chen J, Cheng Q, Liu Y, Li T. The paracrine effect of cobalt chloride on BMSCs during cognitive function rescue in the HIBD rat. Behav Brain Res 2017; 332:99-109. [PMID: 28576310 DOI: 10.1016/j.bbr.2017.05.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 05/20/2017] [Accepted: 05/24/2017] [Indexed: 12/22/2022]
Abstract
Hypoxia-ischemia (HI)-induced perinatal encephalopathy frequently causes chronic neurological morbidities and acute mortality. Bone mesenchymal stem cell (BMSC) transplantation could potentially promote functional and anatomical recovery of ischemic tissue. In vitro hypoxic preconditioning is an effective strategy to improve the survival of BMSCs in ischemic tissue. In this study, cobalt chloride (CoCl2) preconditioned medium from BMSC cultures was injected into the left lateral ventricle of HI rats using a micro-osmotic pump at a flow rate 1.0μl/h for 7 days. The protein levels of HIF-1α and its target genes, vascular endothelial growth factor and erythropoietin, markedly increased after CoCl2 preconditioning in BMSCs. In 7-week-old rats that received CoCl2 preconditioned BMSC medium, results of the Morris water maze test indicated ameliorated spatial working memory function following hypoxia-ischemia damage. Neuronal loss, cellular disorganization, and shrinkage in brain tissue were also ameliorated. Extracellular field excitatory postsynaptic potentials (fEPSPs) in the brain slices of 8-week-old rats were recorded; administration of CoCl2 preconditioned BMSC culture medium induced a progressive increment of baseline and amplitude of the fEPSPs. Immunohistochemical quantification showed that GluR2 protein expression increased. In conclusion, CoCl2 activates HIF-1α signals in BMSCs. CoCl2 preconditioned BMSC culture medium likely effects neuroprotection by inducing long-term potentiation (LTP), which could be associated with GluR2 expression. The paracrine effects of hypoxia preconditioning on BMSCs could have applications in novel cell-based therapeutic strategies for hypoxic and ischemic brain injury.
Collapse
MESH Headings
- Animals
- Brain/pathology
- Brain/physiopathology
- Cells, Cultured
- Cobalt/pharmacology
- Culture Media, Conditioned
- Disease Models, Animal
- Excitatory Postsynaptic Potentials
- Female
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Ischemia, Brain/pathology
- Hypoxia-Ischemia, Brain/physiopathology
- Hypoxia-Ischemia, Brain/psychology
- Hypoxia-Ischemia, Brain/therapy
- Male
- Maze Learning/physiology
- Memory, Short-Term/physiology
- Mesenchymal Stem Cell Transplantation
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/physiology
- Neuroprotection/physiology
- Protective Agents/pharmacology
- Rats, Sprague-Dawley
- Receptors, AMPA/metabolism
- Spatial Memory/physiology
- Tissue Culture Techniques
Collapse
Affiliation(s)
- Ying Dai
- Department of Primary Child Health Care, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Wendi Li
- Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China
| | - Min Zhong
- Department of Neurology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Jie Chen
- Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China
| | - Qian Cheng
- Department of Primary Child Health Care, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Youxue Liu
- Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China.
| | - Tingyu Li
- Department of Primary Child Health Care, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China; Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China.
| |
Collapse
|
47
|
Drommelschmidt K, Serdar M, Bendix I, Herz J, Bertling F, Prager S, Keller M, Ludwig AK, Duhan V, Radtke S, de Miroschedji K, Horn PA, van de Looij Y, Giebel B, Felderhoff-Müser U. Mesenchymal stem cell-derived extracellular vesicles ameliorate inflammation-induced preterm brain injury. Brain Behav Immun 2017; 60:220-232. [PMID: 27847282 DOI: 10.1016/j.bbi.2016.11.011] [Citation(s) in RCA: 211] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 11/04/2016] [Accepted: 11/12/2016] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Preterm brain injury is a major cause of disability in later life, and may result in motor, cognitive and behavioural impairment for which no treatment is currently available. The aetiology is considered as multifactorial, and one underlying key player is inflammation leading to white and grey matter injury. Extracellular vesicles secreted by mesenchymal stem/stromal cells (MSC-EVs) have shown therapeutic potential in regenerative medicine. Here, we investigated the effects of MSC-EV treatment on brain microstructure and maturation, inflammatory processes and long-time outcome in a rodent model of inflammation-induced brain injury. METHODS 3-Day-old Wistar rats (P3) were intraperitoneally injected with 0.25mg/kg lipopolysaccharide or saline and treated with two repetitive doses of 1×108 cell equivalents of MSC-EVs per kg bodyweight. Cellular degeneration and reactive gliosis at P5 and myelination at P11 were evaluated by immunohistochemistry and western blot. Long-term cognitive and motor function was assessed by behavioural testing. Diffusion tensor imaging at P125 evaluated long-term microstructural white matter alterations. RESULTS MSC-EV treatment significantly ameliorated inflammation-induced neuronal cellular degeneration reduced microgliosis and prevented reactive astrogliosis. Short-term myelination deficits and long-term microstructural abnormalities of the white matter were restored by MSC-EV administration. Morphological effects of MSC-EV treatment resulted in improved long-lasting cognitive functions INTERPRETATION: MSC-EVs ameliorate inflammation-induced cellular damage in a rat model of preterm brain injury. MSC-EVs may serve as a novel therapeutic option by prevention of neuronal cell death, restoration of white matter microstructure, reduction of gliosis and long-term functional improvement.
Collapse
Affiliation(s)
- Karla Drommelschmidt
- Department of Paediatrics I/Neonatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Meray Serdar
- Department of Paediatrics I/Neonatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ivo Bendix
- Department of Paediatrics I/Neonatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Josephine Herz
- Department of Paediatrics I/Neonatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Frederik Bertling
- Department of Paediatrics I/Neonatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sebastian Prager
- Department of Paediatrics I/Neonatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Matthias Keller
- Department of Paediatrics I/Neonatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Anna-Kristin Ludwig
- Institute of Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Vikas Duhan
- Institute of Immunology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Stefan Radtke
- Institute of Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; Clinical Research Division, Fred Hutchinson Cancer Research Centre, Seattle, WA 98109, USA
| | - Kyra de Miroschedji
- Institute of Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Peter A Horn
- Institute of Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Yohan van de Looij
- Division of Child Growth and Development, Department of Paediatrics, University of Geneva, Geneva, Switzerland; Laboratory of Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Bernd Giebel
- Institute of Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| | - Ursula Felderhoff-Müser
- Department of Paediatrics I/Neonatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
48
|
Mesenchymal Stromal Cell Therapy for Neonatal Hypoxic-Ischemic Encephalopathy. STEM CELLS IN CLINICAL APPLICATIONS 2017. [DOI: 10.1007/978-3-319-33720-3_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
49
|
Braccioli L, Heijnen CJ, Coffer PJ, Nijboer CH. Delayed administration of neural stem cells after hypoxia-ischemia reduces sensorimotor deficits, cerebral lesion size, and neuroinflammation in neonatal mice. Pediatr Res 2017; 81:127-135. [PMID: 27632779 DOI: 10.1038/pr.2016.172] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/07/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND Hypoxic-ischemic (HI) encephalopathy causes mortality and severe morbidity in neonates. Treatments with a therapeutic window >6 h are currently not available. Here, we explored whether delayed transplantation of allogenic neural stem cells (NSCs) at 10 d after HI could be a tool to repair HI brain injury and improve behavioral impairments. METHODS HI was induced in 9-d-old mice. Animals received NSCs or vehicle intracranially in the hippocampus at 10 d post-HI. Sensorimotor performance was assessed by cylinder rearing test. Lesion size, synaptic integrity, and fate of injected NSCs were determined by immuno-stainings. Neuroinflammation was studied by immuno-stainings of brain sections, primary glial cultures, and TNFα ELISA. RESULTS NSC transplantation at 10 d post-insult induced long-term improvement of motor performance and synaptic integrity, and reduced lesion size compared to vehicle-treatment. HI-induced neuroinflammation was reduced after NSC treatment, at least partially by factors secreted by NSCs. Injected NSCs migrated toward and localized at the damaged hippocampus. Transplanted NSCs differentiated toward the neuronal lineage and formed a niche with endogenous precursors. CONCLUSION Our study provides evidence of the efficacy of NSC transplantation late after HI as a tool to reduce neonatal HI brain injury through regeneration of the lesion.
Collapse
Affiliation(s)
- Luca Braccioli
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht, The Netherlands.,Center for Molecular Medicine and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cobi J Heijnen
- Laboratory of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Paul J Coffer
- Center for Molecular Medicine and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cora H Nijboer
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
50
|
Mueller M, Oppliger B, Joerger-Messerli M, Reinhart U, Barnea E, Paidas M, Kramer BW, Surbek DV, Schoeberlein A. Wharton's Jelly Mesenchymal Stem Cells Protect the Immature Brain in Rats and Modulate Cell Fate. Stem Cells Dev 2016; 26:239-248. [PMID: 27842457 DOI: 10.1089/scd.2016.0108] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The development of a mammalian brain is a complex and long-lasting process. Not surprisingly, preterm birth is the leading cause of death in newborns and children. Advances in perinatal care reduced mortality, but morbidity still represents a major burden. New therapeutic approaches are thus desperately needed. Given that mesenchymal stem/stromal cells (MSCs) emerged as a promising candidate for cell therapy, we transplanted MSCs derived from the Wharton's Jelly (WJ-MSCs) to reduce the burden of immature brain injury in a murine animal model. WJ-MSCs transplantation resulted in protective activity characterized by reduced myelin loss and astroglial activation. WJ-MSCs improved locomotor behavior as well. To address the underlying mechanisms, we tested the key regulators of responses to DNA-damaging agents, such as cyclic AMP-dependent protein kinase/calcium-dependent protein kinase (PKA/PKC), cyclin-dependent kinase (CDK), ataxia-telangiectasia-mutated/ATM- and Rad3-related (ATM/ATR) substrates, protein kinase B (Akt), and 14-3-3 binding protein partners. We characterized WJ-MSCs using a specific profiler polymerase chain reaction array. We provide evidence that WJ-MSCs target pivotal regulators of the cell fate such as CDK/14-3-3/Akt signaling. We identified leukemia inhibitory factor as a potential candidate of WJ-MSCs' induced modifications as well. We hypothesize that WJ-MSCs may exert adaptive responses depending on the type of injury they are facing, making them prominent candidates for cell therapy in perinatal injuries.
Collapse
Affiliation(s)
- Martin Mueller
- 1 Department of Clinical Research, University of Bern , Bern, Switzerland .,2 Department of Obstetrics and Gynecology, University of Bern , Bern, Switzerland .,3 Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine , New Haven, Connecticut
| | - Byron Oppliger
- 1 Department of Clinical Research, University of Bern , Bern, Switzerland .,2 Department of Obstetrics and Gynecology, University of Bern , Bern, Switzerland
| | - Marianne Joerger-Messerli
- 1 Department of Clinical Research, University of Bern , Bern, Switzerland .,2 Department of Obstetrics and Gynecology, University of Bern , Bern, Switzerland
| | - Ursula Reinhart
- 1 Department of Clinical Research, University of Bern , Bern, Switzerland .,2 Department of Obstetrics and Gynecology, University of Bern , Bern, Switzerland
| | - Eytan Barnea
- 4 Society for the Investigation of Early Pregnancy and BioIncept LLC , Cherry Hill, New Jersey
| | - Michael Paidas
- 3 Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine , New Haven, Connecticut
| | - Boris W Kramer
- 5 Department of Pediatrics, Maastricht University Medical Center (MUMC) , Maastricht, the Netherlands .,6 Division Neuroscience, Department of Neuropsychology, School of Mental Health and Neuroscience (MHeNS), Maastricht University , Maastricht, the Netherlands
| | - Daniel V Surbek
- 1 Department of Clinical Research, University of Bern , Bern, Switzerland .,2 Department of Obstetrics and Gynecology, University of Bern , Bern, Switzerland
| | - Andreina Schoeberlein
- 1 Department of Clinical Research, University of Bern , Bern, Switzerland .,2 Department of Obstetrics and Gynecology, University of Bern , Bern, Switzerland
| |
Collapse
|