1
|
Lin TI, Hsieh PY, Lin HJ, Chiang CK, Sheu JJC, Chang WT, Liau I, Hsu HY. Soy Protein-Cultured Mesenchymal Stem Cell-Secreted Extracellular Vesicles Target the Neurovascular Unit: Insights from a Zebrafish Brain Injury Model. ACS Biomater Sci Eng 2025; 11:1432-1444. [PMID: 40000145 PMCID: PMC11897944 DOI: 10.1021/acsbiomaterials.4c02304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 02/27/2025]
Abstract
Cerebral vascular disorders often accompany hypoxia-induced brain injury. In this study, we develop a zebrafish model of hypoxia-induced cerebral vascular injury to replicate the associated phenotypic changes, including cerebrovascular damage, neuronal apoptosis, and neurological dysfunction. We then explored the therapeutic potential of extracellular vesicles derived from Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) cultured on soy protein-coated surfaces. These vesicles demonstrated superior recovery efficacy, especially in restoring the blood-brain barrier integrity and improving neurological function. Our findings suggest that these potent therapeutic extracellular vesicles, easily produced from WJ-MSCs cultured in the presence of soy proteins, may mitigate hypoxia-induced brain injury by decreasing the severity of vascular disorder caused by oxidative stress. Protein-protein interactome analysis further suggests that multiple signaling pathways are likely involved in restoring normal neurovascular unit function.
Collapse
Affiliation(s)
- Tai-I Lin
- Department
of Applied Chemistry and Institute of Molecular Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
| | - Pei-Ying Hsieh
- Department
of Applied Chemistry and Institute of Molecular Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
| | - Hui-Jen Lin
- Department
of Applied Chemistry and Institute of Molecular Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
| | - Cheng-Kang Chiang
- Department
of Chemistry, National Dong Hwa University, Hualien 974301, Taiwan
| | - Jim Jinn-Chyuan Sheu
- Institute
of Biomedical Sciences, National Sun Yat-Sen
University, Kaohsiung 804201, Taiwan
| | - Wei-Tien Chang
- National
Taiwan University Hospital/National Taiwan University, Taipei 100233, Taiwan
| | - Ian Liau
- Department
of Applied Chemistry and Institute of Molecular Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
- Center
for Emergent Functional Matter Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
| | - Hsin-Yun Hsu
- Department
of Applied Chemistry and Institute of Molecular Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
- Center
for Emergent Functional Matter Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
| |
Collapse
|
2
|
Buccilli B. Pediatric stroke: We need to look for it. J Neurol Sci 2024; 467:123276. [PMID: 39510868 DOI: 10.1016/j.jns.2024.123276] [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: 07/21/2023] [Revised: 09/28/2024] [Accepted: 10/14/2024] [Indexed: 11/15/2024]
Abstract
PURPOSE This review provides a comprehensive overview of the characteristics and diagnosis of pediatric stroke, emphasizing the importance of early recognition and accurate assessment. Pediatric stroke is a complex condition with diverse etiologies, and its timely diagnosis is critical for initiating appropriate interventions and improving clinical outcomes. RECENT FINDINGS Recent advances in neuroimaging techniques, including magnetic resonance imaging (MRI) and magnetic resonance angiography (MRA), have significantly enhanced the diagnostic capabilities for pediatric stroke. Additionally, a better understanding of its underlying etiologies in specific cases, and of the importance of differential diagnosis have improved the outcome and prevention strategies in this vulnerable population. Despite these improvements, though, research still has a long way to go to optimize the management of this condition. SUMMARY Timely and accurate diagnosis of pediatric stroke remains a challenge due to its rarity and variability in clinical presentation, and to the presence of many mimic conditions. The integration of clinical evaluation, neuroimaging, and comorbidities analysis is crucial for achieving a precise diagnosis and guiding tailored treatment strategies for affected children.
Collapse
Affiliation(s)
- Barbara Buccilli
- Icahn School of Medicine at Mount Sinai, Department of Neurosurgery, 1 Gustave L. Levy Place, New York, NY 10029-6574, United States of America
| |
Collapse
|
3
|
Penny TR, Jenkin G, Miller SL, McDonald CA. Umbilical cord blood derived cell expansion: a potential neuroprotective therapy. Stem Cell Res Ther 2024; 15:234. [PMID: 39075614 PMCID: PMC11287950 DOI: 10.1186/s13287-024-03830-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 07/02/2024] [Indexed: 07/31/2024] Open
Abstract
Umbilical cord blood (UCB) is a rich source of beneficial stem and progenitor cells with known angiogenic, neuroregenerative and immune-modulatory properties. Preclinical studies have highlighted the benefit of UCB for a broad range of conditions including haematological conditions, metabolic disorders and neurological conditions, however clinical translation of UCB therapies is lacking. One barrier for clinical translation is inadequate cell numbers in some samples meaning that often a therapeutic dose cannot be achieved. This is particularly important when treating adults or when administering repeat doses of cells. To overcome this, UCB cell expansion is being explored to increase cell numbers. The current focus of UCB cell expansion is CD34+ haematopoietic stem cells (HSCs) for which the main application is treatment of haematological conditions. Currently there are 36 registered clinical trials that are examining the efficacy of expanded UCB cells with 31 of these being for haematological malignancies. Early data from these trials suggest that expanded UCB cells are a safe and feasible treatment option and show greater engraftment potential than unexpanded UCB. Outside of the haematology research space, expanded UCB has been trialled as a therapy in only two preclinical studies, one for spinal cord injury and one for hind limb ischemia. Proteomic analysis of expanded UCB cells in these studies showed that the cells were neuroprotective, anti-inflammatory and angiogenic. These findings are also supported by in vitro studies where expanded UCB CD34+ cells showed increased gene expression of neurotrophic and angiogenic factors compared to unexpanded CD34+ cells. Preclinical evidence demonstrates that unexpanded CD34+ cells are a promising therapy for neurological conditions where they have been shown to improve multiple indices of injury in rodent models of stroke, Parkinson's disease and neonatal hypoxic ischemic brain injury. This review will highlight the current application of expanded UCB derived HSCs in transplant medicine, and also explore the potential use of expanded HSCs as a therapy for neurological conditions. It is proposed that expanded UCB derived CD34+ cells are an appropriate cellular therapy for a range of neurological conditions in children and adults.
Collapse
Affiliation(s)
- Tayla R Penny
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia.
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Courtney A McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| |
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
|
Buccilli B. Exploring new horizons: Emerging therapeutic strategies for pediatric stroke. Exp Neurol 2024; 374:114701. [PMID: 38278205 DOI: 10.1016/j.expneurol.2024.114701] [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: 09/29/2023] [Revised: 12/31/2023] [Accepted: 01/23/2024] [Indexed: 01/28/2024]
Abstract
Pediatric stroke presents unique challenges, and optimizing treatment strategies is essential for improving outcomes in this vulnerable population. This review aims to provide an overview of new, innovative, and potential treatments for pediatric stroke, with a primary objective to stimulate further research in this field. Our review highlights several promising approaches in the realm of pediatric stroke management, including but not limited to stem cell therapy and robotic rehabilitation. These innovative interventions offer new avenues for enhancing functional recovery, reducing long-term disability, and tailoring treatments to individual patient needs. The findings of this review underscore the importance of ongoing research and development of innovative treatments in pediatric stroke. These advancements hold significant clinical relevance, offering the potential to improve the lives of children affected by stroke by enhancing the precision, efficacy, and accessibility of therapeutic interventions. Embracing these innovations is essential in our pursuit of better outcomes and a brighter future for pediatric stroke care.
Collapse
Affiliation(s)
- Barbara Buccilli
- Icahn School of Medicine at Mount Sinai, Department of Neurosurgery, 1 Gustave L. Levy Pl, New York, NY 10029, United States of America.
| |
Collapse
|
6
|
Chen XF, Wu Y, Kim B, Nguyen KV, Chen A, Qiu J, Santoso AR, Disdier C, Lim YP, Stonestreet BS. Neuroprotective efficacy of hypothermia and Inter-alpha Inhibitor Proteins after hypoxic ischemic brain injury in neonatal rats. Neurotherapeutics 2024; 21:e00341. [PMID: 38453562 PMCID: PMC11070713 DOI: 10.1016/j.neurot.2024.e00341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/09/2024] Open
Abstract
Therapeutic hypothermia is the standard of care for hypoxic-ischemic (HI) encephalopathy. Inter-alpha Inhibitor Proteins (IAIPs) attenuate brain injury after HI in neonatal rats. Human (h) IAIPs (60 mg/kg) or placebo (PL) were given 15 min, 24 and 48 h to postnatal (P) day-7 rats after carotid ligation and 8% oxygen for 90 min with (30 °C) and without (36 °C) exposure to hypothermia 1.5 h after HI for 3 h. Hemispheric volume atrophy (P14) and neurobehavioral tests including righting reflex (P8-P10), small open field (P13-P14), and negative geotaxis (P14) were determined. Hemispheric volume atrophy in males was reduced (P < 0.05) by 41.9% in the normothermic-IAIP and 28.1% in the hypothermic-IAIP compared with the normothermic-PL group, and in females reduced (P < 0.05) by 30.3% in the normothermic-IAIP, 45.7% in hypothermic-PL, and 55.2% in hypothermic-IAIP compared with the normothermic-PL group after HI. Hypothermia improved (P < 0.05) the neuroprotective effects of hIAIPs in females. The neuroprotective efficacy of hIAIPs was comparable to hypothermia in female rats (P = 0.183). Treatment with hIAIPs, hypothermia, and hIAIPs with hypothermia decreased (P < 0.05) the latency to enter the peripheral zone in the small open field test in males. We conclude that hIAIPs provide neuroprotection from HI brain injury that is comparable to the protection by hypothermia, hypothermia increases the effects of hIAIPs in females, and hIAIPs and hypothermia exhibit some sex-related differential effects.
Collapse
Affiliation(s)
- Xiaodi F Chen
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Alpert Medical School of Brown University, USA
| | - Yuqi Wu
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Alpert Medical School of Brown University, USA
| | - Boram Kim
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Alpert Medical School of Brown University, USA
| | - Kevin V Nguyen
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Alpert Medical School of Brown University, USA
| | - Ainuo Chen
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Alpert Medical School of Brown University, USA
| | - Joseph Qiu
- ProThera Biologics, Inc., Providence, RI, USA
| | | | - Clemence Disdier
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Alpert Medical School of Brown University, USA
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI, USA; The Alpert Medical School of Brown University, Department of Pathology and Laboratory Medicine, Providence, RI, USA
| | - Barbara S Stonestreet
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Alpert Medical School of Brown University, USA; Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA.
| |
Collapse
|
7
|
Di Martino E, Rayasam A, Vexler ZS. Brain Maturation as a Fundamental Factor in Immune-Neurovascular Interactions in Stroke. Transl Stroke Res 2024; 15:69-86. [PMID: 36705821 PMCID: PMC10796425 DOI: 10.1007/s12975-022-01111-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 01/28/2023]
Abstract
Injuries in the developing brain cause significant long-term neurological deficits. Emerging clinical and preclinical data have demonstrated that the pathophysiology of neonatal and childhood stroke share similar mechanisms that regulate brain damage, but also have distinct molecular signatures and cellular pathways. The focus of this review is on two different diseases-neonatal and childhood stroke-with emphasis on similarities and distinctions identified thus far in rodent models of these diseases. This includes the susceptibility of distinct cell types to brain injury with particular emphasis on the role of resident and peripheral immune populations in modulating stroke outcome. Furthermore, we discuss some of the most recent and relevant findings in relation to the immune-neurovascular crosstalk and how the influence of inflammatory mediators is dependent on specific brain maturation stages. Finally, we comment on the current state of treatments geared toward inducing neuroprotection and promoting brain repair after injury and highlight that future prophylactic and therapeutic strategies for stroke should be age-specific and consider gender differences in order to achieve optimal translational success.
Collapse
Affiliation(s)
- Elena Di Martino
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Aditya Rayasam
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Zinaida S Vexler
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA.
| |
Collapse
|
8
|
Mallard C, Ferriero DM, Vexler ZS. Immune-Neurovascular Interactions in Experimental Perinatal and Childhood Arterial Ischemic Stroke. Stroke 2024; 55:506-518. [PMID: 38252757 DOI: 10.1161/strokeaha.123.043399] [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] [Indexed: 01/24/2024]
Abstract
Emerging clinical and preclinical data have demonstrated that the pathophysiology of arterial ischemic stroke in the adult, neonates, and children share similar mechanisms that regulate brain damage but also have distinct molecular signatures and involved cellular pathways due to the maturational stage of the central nervous system and the immune system at the time of the insult. In this review, we discuss similarities and differences identified thus far in rodent models of 2 different diseases-neonatal (perinatal) and childhood arterial ischemic stroke. In particular, we review acquired knowledge of the role of resident and peripheral immune populations in modulating outcomes in models of perinatal and childhood arterial ischemic stroke and the most recent and relevant findings in relation to the immune-neurovascular crosstalk, and how the influence of inflammatory mediators is dependent on specific brain maturation stages. Finally, we discuss the current state of treatments geared toward age-appropriate therapies that signal via the immune-neurovascular interaction and consider sex differences to achieve successful translation.
Collapse
Affiliation(s)
- Carina Mallard
- Department of Physiology, Institute of Neuroscience and Physiology, University of Gothenburg, Sweden (C.M.)
| | - Donna M Ferriero
- Department of Pediatrics, UCSF, San Francisco, CA (D.M.F.)
- Department of Neurology, UCSF, Weill Institute for Neurosciences, San Francisco, CA (D.M.F., Z.S.V.)
| | - Zinaida S Vexler
- Department of Neurology, UCSF, Weill Institute for Neurosciences, San Francisco, CA (D.M.F., Z.S.V.)
| |
Collapse
|
9
|
Scrutton AM, Ollis F, Boltze J. Mononuclear cell therapy of neonatal hypoxic-ischemic encephalopathy in preclinical versus clinical studies: a systematic analysis of therapeutic efficacy and study design. NEUROPROTECTION 2023; 1:143-159. [PMID: 38213793 PMCID: PMC7615506 DOI: 10.1002/nep3.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/08/2023] [Indexed: 01/13/2024]
Abstract
Background Hypoxic-ischemic encephalopathy (HIE) is a devastating condition affecting around 8.5 in 1000 newborns globally. Therapeutic hypothermia (TH) can reduce mortality and, to a limited extent, disability after HIE. Nevertheless, there is a need for new and effective treatment strategies. Cell based treatments using mononuclear cells (MNC), which can be sourced from umbilical cord blood, are currently being investigated. Despite promising preclinical results, there is currently no strong indicator for clinical efficacy of the approach. This analysis aimed to provide potential explanations for this discrepancy. Methods A systematic review and meta-analysis was conducted according to the Preferred Reporting Items for Systematic reviews and Meta-Analysis (PRISMA) guidelines. Preclinical and clinical studies were retrieved from PubMed, Web of Science, Scopus, and clinicaltrials.gov using a predefined search strategy. A total of 17 preclinical and 7 clinical studies were included. We analyzed overall MNC efficacy in preclinical trials, the methodological quality of preclinical trials and relevant design features in preclinical versus clinical trials. Results There was evidence for MNC therapeutic efficacy in preclinical models of HIE. The methodological quality of preclinical studies was not optimal, and statistical design quality was particularly poor. However, methodological quality was above the standard in other fields. There were significant differences in preclinical versus clinical study design including the use of TH as a baseline treatment (only in clinical studies) and much higher MNC doses being applied in preclinical studies. Conclusions Based on the analyzed data, it is unlikely that therapeutic effect size is massively overestimated in preclinical studies. It is more plausible that the many design differences between preclinical and clinical trials are responsible for the so far lacking proof of efficacy of MNC treatments in HIE. Additional preclinical and clinical research is required to optimize the application of MNC for experimental HIE treatment.
Collapse
Affiliation(s)
- Alexander M. Scrutton
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
- Neurobiology Division, MRC Laboratory of Molecular Biology, University of Cambridge, Cambridge, United Kingdom
| | - Francesca Ollis
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
10
|
Zhou J, Gao T, Tang W, Qian T, Wang Z, Xu P, Wang L. Progress in the treatment of neonatal hypoxic-ischemic encephalopathy with umbilical cord blood mononuclear cells. Brain Dev 2023; 45:533-546. [PMID: 37806836 DOI: 10.1016/j.braindev.2023.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/05/2023] [Accepted: 09/11/2023] [Indexed: 10/10/2023]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is a common disease among newborns, which is a leading cause of neonatal death and permanent neurological sequelae. Therapeutic hypothermia (TH) is the only method for the treatment of HIE that has been recognized effective clinically at home and abroad, but the efficacy is limited. Recent research suggests that the cord blood-derived mononuclear cells (CB-MNCs), which the refer to blood cells containing one nucleus in the cord blood, exert anti-oxidative, anti-inflammatory, anti-apoptotic effects and play a neuroprotective role in HIE. This review focuses on safety and efficacy, the route of administration, dose, timing and combination treatment of CB-MNCs in HIE.
Collapse
Affiliation(s)
- Jiayu Zhou
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, China
| | - Ting Gao
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, China
| | - Wan Tang
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, China
| | - Tianyang Qian
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, China
| | - Ziming Wang
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, China
| | - Pu Xu
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, China
| | - Laishuan Wang
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, China.
| |
Collapse
|
11
|
Wang Z, Zhang L, Yang Y, Wang Q, Qu S, Wang X, He Z, Luan Z. Oligodendrocyte Progenitor Cell Transplantation Ameliorates Preterm Infant Cerebral White Matter Injury in Rats Model. Neuropsychiatr Dis Treat 2023; 19:1935-1947. [PMID: 37719062 PMCID: PMC10503552 DOI: 10.2147/ndt.s414493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/24/2023] [Indexed: 09/19/2023] Open
Abstract
Background Cerebral white matter injury (WMI) is the most common brain injury in preterm infants, leading to motor and developmental deficits often accompanied by cognitive impairment. However, there is no effective treatment. One promising approach for treating preterm WMI is cell replacement therapy, in which lost cells can be replaced by exogenous oligodendrocyte progenitor cells (OPCs). Methods This study developed a method to differentiate human neural stem cells (hNSCs) into human OPCs (hOPCs). The preterm WMI animal model was established in rats on postnatal day 3, and OLIG2+/NG2+/PDGFRα+/O4+ hOPCs were enriched and transplanted into the corpus callosum on postnatal day 10. Then, histological analysis and electron microscopy were used to detect lesion structure; behavioral assays were performed to detect cognitive function. Results Transplanted hOPCs survived and migrated throughout the major white matter tracts. Morphological differentiation of transplanted hOPCs was observed. Histological analysis revealed structural repair of lesioned areas. Re-myelination of the axons in the corpus callosum was confirmed by electron microscopy. The Morris water maze test revealed cognitive function recovery. Conclusion Our study showed that exogenous hOPCs could differentiate into CC1+ OLS in the brain of WMI rats, improving their cognitive functions.
Collapse
Affiliation(s)
- Zhaoyan Wang
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, People’s Republic of China
| | - Leping Zhang
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, People’s Republic of China
- Guizhou Medical University, Guiyang, 550004, People’s Republic of China
| | - Yinxiang Yang
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, People’s Republic of China
| | - Qian Wang
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, People’s Republic of China
| | - Suqing Qu
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, People’s Republic of China
| | - Xiaohua Wang
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, People’s Republic of China
| | - Zhixu He
- Guizhou Medical University, Guiyang, 550004, People’s Republic of China
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563100, People’s Republic of China
| | - Zuo Luan
- Laboratory of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, People’s Republic of China
| |
Collapse
|
12
|
Pimentel‐Coelho PM. Monocytes in neonatal stroke and hypoxic‐ischemic encephalopathy: Pathophysiological mechanisms and therapeutic possibilities. NEUROPROTECTION 2023; 1:66-79. [DOI: 10.1002/nep3.22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/13/2023] [Indexed: 01/03/2025]
Abstract
AbstractNeonatal arterial ischemic stroke (NAIS) and neonatal hypoxic‐ischemic encephalopathy (HIE) are common causes of neurological impairments in infants, for which treatment options are very limited. NAIS and HIE induce an innate immune response that involves the recruitment of peripheral immune cells, including monocytes, into the brain. Monocytes and monocyte‐derived cells have the potential to contribute to both harmful and beneficial pathophysiological processes, such as neuroinflammation and brain repair, but their roles in NAIS and HIE remain poorly understood. Furthermore, recent evidence indicates that monocyte‐derived macrophages can persist in the brain for several months following NAIS and HIE in mice, with possible long‐lasting consequences that are still unknown. This review provides a comprehensive overview of the mechanisms of monocyte infiltration and their potential functions in the ischemic brain, focusing on HIE and NAIS. Therapeutic strategies targeting monocytes and the possibility of using monocytes for cell‐based therapies are also discussed.
Collapse
Affiliation(s)
- Pedro M. Pimentel‐Coelho
- Carlos Chagas Filho Biophysics Institute Federal University of Rio de Janeiro Rio de Janeiro Brazil
| |
Collapse
|
13
|
Nguyen T, Purcell E, Smith MJ, Penny TR, Paton MCB, Zhou L, Jenkin G, Miller SL, McDonald CA, Malhotra A. Umbilical Cord Blood-Derived Cell Therapy for Perinatal Brain Injury: A Systematic Review & Meta-Analysis of Preclinical Studies. Int J Mol Sci 2023; 24:ijms24054351. [PMID: 36901781 PMCID: PMC10001969 DOI: 10.3390/ijms24054351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/12/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Perinatal brain injury is a major contributor to long-term adverse neurodevelopment. There is mounting preclinical evidence for use of umbilical cord blood (UCB)-derived cell therapy as potential treatment. To systematically review and analyse effects of UCB-derived cell therapy on brain outcomes in preclinical models of perinatal brain injury. MEDLINE and Embase databases were searched for relevant studies. Brain injury outcomes were extracted for meta-analysis to calculate standard mean difference (SMD) with 95% confidence interval (CI), using an inverse variance, random effects model. Outcomes were separated based on grey matter (GM) and white matter (WM) regions where applicable. Risk of bias was assessed using SYRCLE, and GRADE was used to summarise certainty of evidence. Fifty-five eligible studies were included (7 large, 48 small animal models). UCB-derived cell therapy significantly improved outcomes across multiple domains, including decreased infarct size (SMD 0.53; 95% CI (0.32, 0.74), p < 0.00001), apoptosis (WM, SMD 1.59; 95%CI (0.86, 2.32), p < 0.0001), astrogliosis (GM, SMD 0.56; 95% CI (0.12, 1.01), p = 0.01), microglial activation (WM, SMD 1.03; 95% CI (0.40, 1.66), p = 0.001), neuroinflammation (TNF-α, SMD 0.84; 95%CI (0.44, 1.25), p < 0.0001); as well as improved neuron number (SMD 0.86; 95% CI (0.39, 1.33), p = 0.0003), oligodendrocyte number (GM, SMD 3.35; 95 %CI (1.00, 5.69), p = 0.005) and motor function (cylinder test, SMD 0.49; 95 %CI (0.23, 0.76), p = 0.0003). Risk of bias was determined as serious, and overall certainty of evidence was low. UCB-derived cell therapy is an efficacious treatment in pre-clinical models of perinatal brain injury, however findings are limited by low certainty of evidence.
Collapse
Affiliation(s)
- Timothy Nguyen
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| | - Elisha Purcell
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| | - Madeleine J. Smith
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC 3168, Australia
| | - Tayla R. Penny
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC 3168, Australia
| | - Madison C. B. Paton
- Cerebral Palsy Alliance Research Institute & Specialty of Child and Adolescent Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Lindsay Zhou
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC 3168, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC 3168, Australia
| | - Suzanne L. Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC 3168, Australia
| | - Courtney A. McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC 3168, Australia
| | - Atul Malhotra
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC 3168, Australia
- Correspondence:
| |
Collapse
|
14
|
Bell A, Watt AP, Dudink I, Pham Y, Sutherland AE, Allison BJ, McDonald CA, Castillo-Melendez M, Jenkin G, Malhotra A, Miller SL, Yawno T. Endothelial colony forming cell administration promotes neurovascular unit development in growth restricted and appropriately grown fetal lambs. Stem Cell Res Ther 2023; 14:29. [PMID: 36788590 PMCID: PMC9930266 DOI: 10.1186/s13287-023-03249-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 02/01/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Fetal growth restriction (FGR) is associated with deficits in the developing brain, including neurovascular unit (NVU) dysfunction. Endothelial colony forming cells (ECFC) can mediate improved vascular stability, and have demonstrated potential to enhance vascular development and protection. This investigation examined whether ECFCs from human umbilical cord blood (UCB) enhanced NVU development in FGR and appropriate for gestational age (AGA) fetal sheep. METHODS Twin-bearing ewes had surgery performed at 88-90 days' gestation, inducing FGR in one fetus. At 113 days, ECFCs (1 × 107 cells) cultured from human UCB were administered intravenously to fetal sheep in utero. At 127 days, ewes and their fetuses were euthanised, fetal brains collected, and NVU components analysed by immunohistochemistry. RESULTS Twenty-four fetal lambs, arranged in four groups: AGA (n = 7), FGR (n = 5), AGA + ECFC (n = 6), and FGR + ECFC (n = 6), were included in analyses. FGR resulted in lower body weight than AGA (P = 0.002) with higher brain/body weight ratio (P = 0.003). ECFC treatment was associated with increased vascular density throughout the brain in both AGA + ECFC and FGR + ECFC groups, as well as increased vascular-astrocyte coverage and VEGF expression in the cortex (P = 0.003, P = 0.0006, respectively) and in the subcortical white matter (P = 0.01, P = 0.0002, respectively) when compared with the untreated groups. CONCLUSIONS ECFC administration enhanced development of NVU components in both the AGA and FGR fetal brain. Further investigation is required to assess how to optimise the enhanced angiogenic capabilities of ECFCs to provide a therapeutic strategy to protect the developing NVU against vulnerabilities associated with FGR.
Collapse
Affiliation(s)
- Alexander Bell
- grid.452824.dThe Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia ,grid.1002.30000 0004 1936 7857Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Ashalyn P. Watt
- grid.452824.dThe Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia
| | - Ingrid Dudink
- grid.452824.dThe Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia ,grid.1002.30000 0004 1936 7857Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Yen Pham
- grid.452824.dThe Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia
| | - Amy E. Sutherland
- grid.452824.dThe Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia
| | - Beth J. Allison
- grid.452824.dThe Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia ,grid.1002.30000 0004 1936 7857Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Courtney A. McDonald
- grid.452824.dThe Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia ,grid.1002.30000 0004 1936 7857Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | | | - Graham Jenkin
- grid.452824.dThe Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia ,grid.1002.30000 0004 1936 7857Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Atul Malhotra
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia. .,Department of Paediatrics, Monash University, 246 Clayton Road, Clayton, Melbourne, VIC, 3168, Australia. .,Monash Newborn, Monash Children's Hospital, Melbourne, Australia.
| | - Suzanne L. Miller
- grid.452824.dThe Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia ,grid.1002.30000 0004 1936 7857Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Tamara Yawno
- grid.452824.dThe Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia ,grid.1002.30000 0004 1936 7857Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia ,grid.1002.30000 0004 1936 7857Department of Paediatrics, Monash University, 246 Clayton Road, Clayton, Melbourne, VIC 3168 Australia
| |
Collapse
|
15
|
Ovcjak A, Pontello R, Miller SP, Sun HS, Feng ZP. Hypothermia combined with neuroprotective adjuvants shortens the duration of hospitalization in infants with hypoxic ischemic encephalopathy: Meta-analysis. Front Pharmacol 2023; 13:1037131. [PMID: 36686686 PMCID: PMC9853207 DOI: 10.3389/fphar.2022.1037131] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/20/2022] [Indexed: 01/09/2023] Open
Abstract
Objective: Therapeutic hypothermia (TH) is the current standard of care for neonatal hypoxic-ischemic encephalopathy (HIE), yet morbidity and mortality remain significant. Adjuvant neuroprotective agents have been suggested to augment hypothermic-mediated neuroprotection. This analysis aims to identify the classes of drugs that have been used in combination with hypothermia in the treatment of neonatal HIE and determine whether combination therapy is more efficacious than TH alone. Methods: A systematic search of PubMed, Embase and Medline from conception through December 2022 was conducted. Randomized- and quasi-randomized controlled trials, observational studies and retrospective studies evaluating HIE infants treated with combination therapy versus TH alone were selected. Primary reviewers extracted information on mortality, neurodevelopmental impairment and length of hospitalization for meta-analyses. Effect sizes were pooled using a random-effects model and measured as odds ratio (OR) or mean difference (MD) where applicable, and 95% confidence intervals (CI) were calculated. Risk of bias was assessed using the tool from the Cochrane Handbook for Systematic Reviews of Interventions. Results: The search strategy collected 519 studies, 16 of which met analysis inclusion criteria. HIE infants totaled 1,288 infants from included studies, 646 infants received some form of combination therapy, while 642 received TH alone. GABA receptor agonists, NMDA receptor antagonists, neurogenic and angiogenic agents, stem cells, glucocorticoids and antioxidants were identified as candidate adjuvants to TH that have been evaluated in clinical settings compared to TH alone. Length of hospitalization was significantly reduced in infants treated with combination therapy (MD -4.81, 95% CI [-8.42. to -1.19], p = .009) compared to those treated with TH alone. Risk of mortality and neurodevelopmental impairment did not differ between combination therapy and TH alone groups. Conclusion: Compared to the current standard of care, administration of neuroprotective adjuvants with TH reduced the duration of hospitalization but did not impact the risk of mortality or neurodevelopmental impairment in HIE infants. Meta-analysis was limited by a moderate risk of bias among included studies and small sample sizes. This analysis highlights the need for preclinical trials to conduct drug development studies in hypothermic settings to identify relevant molecular targets that may offer additive or synergistic neuroprotection to TH, and the need for larger powered clinical trials to determine the dose and timing of administration at which maximal clinical benefits are observed for adjuvant neuroprotectants.
Collapse
Affiliation(s)
- Andrea Ovcjak
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Riley Pontello
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Steve P. Miller
- Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, Faculty of Medicine, The University of Toronto, Toronto, ON, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Zhong-Ping Feng
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
16
|
Yousif NG, Yousif MG, Mohsen AAU, El-Bakaa HS, Younise MH, Altimimi AN, Nöth UA, Hassan AM. PROSPECTIVE SINGLE CENTER ANALYSIS OF OUTCOME STEM CELLS TRANSPLANTS IN PATIENTS WITH CEREBRAL PALSY. POLSKI MERKURIUSZ LEKARSKI : ORGAN POLSKIEGO TOWARZYSTWA LEKARSKIEGO 2023; 51:339-345. [PMID: 37756453 DOI: 10.36740/merkur202304107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
OBJECTIVE Aim: To evaluate efficacy and safety of autologous bone marrow-derived mononuclear stem cell transplantation intrathecal in children with cerebral palsy. PATIENTS AND METHODS Materials and Methods: 35 children have levels I-V cerebral palsy aged 8-months to 8-years-old were enrolled from September (2021-2022) at Iraqi private hospital. Gross Motor Function was assessed by a pediatrician and neurologist specialist, 5 mcg/kg/day of G-CSF subcutaneous single injection daily for three consecutive days. Bone marrow harvested from posterior iliac crest under light general anesthesia. Bone marrow mononuclear cells (BMMNCs) separation was performed using density gradient centrifugation with Ficoll, the cell viability checked by propidium iodide dye in a TALI machine (Invitrogen) in average 98%. The viable BMMNCs injected intrathecal in L4-L5 over a period of 5-10 min. RESULTS Results: Males accounted for 57.14% (20/35) while female 42.86% (15/35), and main neurological symptoms included spastic disorder spastic disorder (quadriplegia 24 (68.6), tetraplegia 2 (5.7), diplegia 5 (14.28), hemiplegia4 (11.42)). Gross Motor Function Classification System and Gross Motor Function Measure-66 (GMFM-66) showed II 10 (28.58), III 11(31.42) and IV 14 (40). On mean follow-up of 3 months post-stem cell transplant improvement was observed in 80% cases. The improvement showed in gross motor function (6/8) p=0.01, and speech (2/4) p=0.04, neck holding (5/5) p=0.0003, sitting balance (4/4) p=0.04, postural tone (5/5) p=0.0003, as well as significant reduction in seizure frequency (2/3) p=0.04 and improvement in cognition (6/7) p=0.01 were observed. CONCLUSION Conclusion: Stem cell therapy for cerebral palsy shows a significant positive effect on the gross motor function, without long adverse effects.
Collapse
Affiliation(s)
- Nasser Ghaly Yousif
- DEPARTMENT OF MEDICINE, MEDICAL COLLEGE, AL MUTHANNA UNIVERSITY, SAMAWAH, IRAQ
| | - Maitham G Yousif
- DEPARTMENT OF BIOLOGY, COLLEGE OF SCIENCE, AL-QADISIYAH UNIVERSITY, IRAQ
| | - Ahmed Abd Ulhadi Mohsen
- DEPARTMENT OF PEDIATRIC, COLLEGE OF MEDICINE, JABIR IBN HAYYAN MEDICAL UNIVERSITY, KUFA, IRAQ
| | | | | | - Ahmed N Altimimi
- DEPARTMENT OF BIOLOGY, ALHAKEEM HOSPITAL, MINISTRY OF HEALTH, AL NAJAF, IRAQ
| | - Ulrich Aran Nöth
- DEPARTMENT OF REGENERATIVE RESEARCH, COLLEGE OF MEDICINE, COLORADO UNIVERSITY, AURORA, USA
| | - Alaa Manea Hassan
- DEPARTMENT OF DRUG CONTROL AND RESEARCH LABORATORY (DCRL), MINISTRY OF HEALTH, AL NAJAF, IRAQ
| |
Collapse
|
17
|
Rao RB, Shiao ML, Ennis-Czerniak KM, Low WC. Nonhematopoietic Umbilical Cord Blood Stem Cell Administration Improves Long-term Neurodevelopment After Periventricular-Intraventricular Hemorrhage in Neonatal Rats. Cell Transplant 2023; 32:9636897231189301. [PMID: 37493283 PMCID: PMC10387682 DOI: 10.1177/09636897231189301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 06/27/2023] [Accepted: 07/05/2023] [Indexed: 07/27/2023] Open
Abstract
Periventricular-intraventricular hemorrhage (PIVH) is common in extremely low gestational age neonates (ELGAN) and leads to motor and behavioral impairments. Currently there is no effective treatment for PIVH. Whether human nonhematopoietic umbilical cord blood-derived stem cell (nh-UCBSC) administration reduces the severity of brain injury and improves long-term motor and behavioral function was tested in an ELGAN-equivalent neonatal rat model of PIVH. In a collagenase-induced unilateral PIVH on postnatal day (P) 2 model, rat pups received a single dose of nh-UCBSCs at a dose of 1 × 106 cells i.p. on P6 (PIVH + UCBSC group) or were left untreated (Untreated PIVH group). Motor deficit was determined using forelimb placement, edge-push, and elevated body swing tests at 2 months (N = 5-8). Behavior was evaluated using open field exploration and rearing tests at 4 months (N =10-12). Cavity volume and hemispheric volume loss on the PIVH side were determined at 7 months (N = 6-7). Outcomes were compared between the Untreated PIVH and PIVH + UCBSC groups and a Control group. Unilateral motor deficits were present in 60%-100% of rats in the Untreated PIVH group and 12.5% rats in the PIVH + UCBSC group (P = 0.02). Untreated PIVH group exhibited a higher number of quadrant crossings in open field exploration, indicating low emotionality and poor habituation, and had a cavitary lesion and hemispheric volume loss on the PIVH side. Performance in open field exploration correlated with cavity volume (r2 = 0.25; P < 0.05). Compared with the Untreated PIVH group, performance in open field exploration was better (P = 0.0025) and hemispheric volume loss was lower (19.9 ± 4.4% vs 6.1 ± 2.6%, P = 0.018) in the PIVH + UCBSC group. These results suggest that a single dose of nh-UCBSCs administered in the subacute period after PIVH reduces the severity of injury and improves neurodevelopment in neonatal rats.
Collapse
Affiliation(s)
- Raghavendra B. Rao
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, MN, USA
| | - Maple L. Shiao
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Kathleen M. Ennis-Czerniak
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Walter C. Low
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
18
|
Serrenho I, Cardoso CM, Grãos M, Dinis A, Manadas B, Baltazar G. Hypothermia Does Not Boost the Neuroprotection Promoted by Umbilical Cord Blood Cells in a Neonatal Hypoxia-Ischemia Rat Model. Int J Mol Sci 2022; 24:ijms24010257. [PMID: 36613698 PMCID: PMC9820288 DOI: 10.3390/ijms24010257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/13/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is one of the leading causes of death and long-term disability in the perinatal period. Currently, therapeutic hypothermia is the standard of care for this condition with modest efficacy and strict enrollment criteria. Therapy with umbilical cord blood cells (UCBC) has come forward as a strong candidate for the treatment of neonatal HIE, but no preclinical studies have yet compared the action of UCBC combined with hypothermia (HT) with the action of each therapy by itself. Thus, to evaluate the potential of each therapeutic approach, a hypoxic-ischemic brain lesion was induced in postnatal day ten rat pups; two hours later, HT was applied for 4 h; and 24, 48, and 72 h post-injury, UCBC were administered intravenously. The neonatal hypoxic-ischemic injury led to a brain lesion involving about 48% of the left hemisphere that was not improved by HT (36%) or UCBC alone (28%), but only with the combined therapies (25%; p = 0.0294). Moreover, a decrease in glial reactivity and improved functional outcomes were observed in both groups treated with UCBC. Overall, these results support UCBC as a successful therapeutic approach for HIE, even when treatment with therapeutic hypothermia is not possible.
Collapse
Affiliation(s)
- Inês Serrenho
- Health Sciences Research Center (CICS-UBI), University of Beira Interior, 6201-506 Covilhã, Portugal
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | | | - Mário Grãos
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Biocant, Technology Transfer Association, 3060-197 Cantanhede, Portugal
- Institute for Interdisciplinary Research, University of Coimbra (IIIUC), 3030-789 Coimbra, Portugal
| | - Alexandra Dinis
- Pediatric Intensive Care Unit, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal
| | - Bruno Manadas
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra (IIIUC), 3030-789 Coimbra, Portugal
- Correspondence: (B.M.); (G.B.)
| | - Graça Baltazar
- Health Sciences Research Center (CICS-UBI), University of Beira Interior, 6201-506 Covilhã, Portugal
- Faculty of Health Sciences, University of Beira Interior, 6201-506 Covilhã, Portugal
- Correspondence: (B.M.); (G.B.)
| |
Collapse
|
19
|
Crompton K, Godler DE, Ling L, Elwood N, Mechinaud-Heloury F, Soosay Raj T, Hsiao KC, Fleming J, Tiedemann K, Novak I, Fahey M, Wang X, Lee KJ, Colditz PB, Edwards P, Reddihough D. Umbilical Cord Blood Cell Clearance Post-Infusion in Immune-Competent Children with Cerebral Palsy. Cells Tissues Organs 2022; 212:546-553. [PMID: 36261026 DOI: 10.1159/000527612] [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: 07/19/2022] [Accepted: 09/13/2022] [Indexed: 11/19/2022] Open
Abstract
Umbilical cord blood cells have therapeutic potential for neurological disorders, through a paracrine mechanism of action. A greater understanding of the safety and immunological effects of allogeneic donor cord blood cells in the context of a healthy recipient immune system, such as in cerebral palsy, is needed. This study aimed to determine how quickly donor cord blood cells were cleared from the circulation in children with cerebral palsy who received a single intravenous infusion of 12/12 human leucocyte antigen (HLA)-matched sibling cord blood cells. Twelve participants with cerebral palsy aged 2-12 years received cord blood cell infusions as part of a phase I trial of umbilical blood infusion for cerebral palsy. Digital droplet PCR analysis of DNA copy number variants specific to donor and recipient was used to assess donor DNA clearance at five timepoints post-infusion, a surrogate measure of cell clearance. Donor cells were cleared by 3 months post-infusion in 11/12 participants. When detected, donor DNA was at a fraction of 0.01-0.31% of total DNA with no signs of graft-versus-host disease in any participant. The donor DNA clearance times provided by this study have important implications for understanding the safety of allogeneic cord blood cell infusion for cerebral palsy and translational tissue engineering or regenerative medicine research in other disorders.
Collapse
Affiliation(s)
- Kylie Crompton
- Neurodisability and Rehabilitation, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Neurodevelopment and Disability, The Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - David E Godler
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Diagnosis and Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Ling Ling
- Diagnosis and Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Ngaire Elwood
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Blood Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- The Royal Children's Hospital, Parkville, Victoria, Australia
- BMDI Cord Blood Bank, Parkville, Victoria, Australia
| | | | - Trisha Soosay Raj
- Children's Cancer Centre, The Royal Children's Hospital, Parkville, Victoria, Australia
- Oncology, Queensland Children's Hospital, South Brisbane, Queensland, Australia
| | - Kuang-Chih Hsiao
- Allergy Immunology, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Immunology, Starship Children's Hospital, Auckland, New Zealand
- Paediatrics, University of Auckland, Auckland, New Zealand
| | - Jacqueline Fleming
- Children's Cancer Centre, The Royal Children's Hospital, Parkville, Victoria, Australia
| | | | - Iona Novak
- Cerebral Palsy Alliance Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Michael Fahey
- Paediatric Neurology, Monash Children's Hospital, Clayton, Victoria, Australia
- Medicine, Monash University, Melbourne, Victoria, Australia
| | - Xiaofang Wang
- Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Katherine J Lee
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Paul B Colditz
- Grantley Stable Neonatal Unit, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
- Perinatal Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Priya Edwards
- Queensland Paediatric Rehabilitation Service, Queensland Children's Hospital, South Brisbane, Queensland, Australia
- Queensland Cerebral Palsy and Rehabilitation Research Centre, The Univeristy of Queensland, Brisbane, Queensland, Australia
| | - Dinah Reddihough
- Neurodisability and Rehabilitation, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Neurodevelopment and Disability, The Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
20
|
Xi Y, Yue G, Gao S, Ju R, Wang Y. Human umbilical cord blood mononuclear cells transplantation for perinatal brain injury. Stem Cell Res Ther 2022; 13:458. [PMID: 36064459 PMCID: PMC9446746 DOI: 10.1186/s13287-022-03153-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/22/2022] [Indexed: 01/06/2023] Open
Abstract
Perinatal brain injury is a leading cause of death and disability in children. Hypoxic-ischemic encephalopathy in full term infants, and white matter injury in premature infants are most known brain injury in perinatal period. Human umbilical cord blood mononuclear cells contain hematopoietic stem cells, mesenchymal stem cells, endothelial progenitor cells, lymphocytes, monocytes, and so on. Human umbilical cord blood mononuclear cells have many biological functions, such as nerve and vascular regeneration, anti-apoptosis, anti-inflammation, and immune regulation. Human umbilical cord blood mononuclear cells transplantation has achieved significant efficacy and safety in animal and clinical trials for the treatment of perinatal brain injury. We will review human umbilical cord blood mononuclear cells transplantation for perinatal brain injury in this review.
Collapse
Affiliation(s)
- Yufeng Xi
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Guang Yue
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Shuqiang Gao
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Rong Ju
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| | - Yujia Wang
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China. .,Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
21
|
Zdolińska-Malinowska I, Boruczkowski D, Hołowaty D, Krajewski P, Snarski E. Rationale for the Use of Cord Blood in Hypoxic-Ischaemic Encephalopathy. Stem Cells Int 2022; 2022:9125460. [PMID: 35599846 PMCID: PMC9117076 DOI: 10.1155/2022/9125460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 02/16/2022] [Accepted: 04/22/2022] [Indexed: 11/18/2022] Open
Abstract
Hypoxic-ischaemic encephalopathy (HIE) is a severe complication of asphyxia at birth. Therapeutic hypothermia, the standard method for HIE prevention, is effective in only 50% of the cases. As the understanding of the immunological basis of these changes increases, experiments have begun with the use of cord blood (CB) because of its neuroprotective properties. Mechanisms for the neuroprotective effects of CB stem cells include antiapoptotic and anti-inflammatory actions, stimulation of angiogenesis, production of trophic factors, and mitochondrial donation. In several animal models of HIE, CB decreased oxidative stress, cell death markers, CD4+ T cell infiltration, and microglial activation; restored normal brain metabolic activity; promoted neurogenesis; improved myelination; and increased the proportion of mature oligodendrocytes, neuron numbers in the motor cortex and somatosensory cortex, and brain weight. These observations translate into motor strength, limb function, gait, and cognitive function and behaviour. In humans, the efficacy and safety of CB administration were reported in a few early clinical studies which confirmed the feasibility and safety of this intervention for up to 10 years. The results of these studies showed an improvement in the developmental outcomes over hypothermia. Two phase-2 clinical studies are ongoing under the United States regulations, namely one controlled study and one blinded study.
Collapse
Affiliation(s)
| | - Dariusz Boruczkowski
- Polski Bank Komórek Macierzystych S.A. (FamiCord Group), Jana Pawła II 29, 00-86 Warsaw, Poland
| | - Dominika Hołowaty
- Department of Obstetrics and Gynecology, Medical University of Warsaw, Starynkiewicza Square 1/3, 02-015 Warsaw, Poland
| | - Paweł Krajewski
- Department of Obstetrics and Gynecology, Medical University of Warsaw, Starynkiewicza Square 1/3, 02-015 Warsaw, Poland
| | - Emilian Snarski
- Polski Bank Komórek Macierzystych S.A. (FamiCord Group), Jana Pawła II 29, 00-86 Warsaw, Poland
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
22
|
Sayin ES, Sobczyk O, Poublanc J, Mikulis DJ, Fisher JA, Kuo KHM, Duffin J. Assessing Cerebrovascular Resistance in Patients With Sickle Cell Disease. Front Physiol 2022; 13:847969. [PMID: 35422710 PMCID: PMC9002264 DOI: 10.3389/fphys.2022.847969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/08/2022] [Indexed: 02/05/2023] Open
Abstract
In patients with sickle cell disease (SCD) the delivery of oxygen to the brain is compromised by anemia, abnormal rheology, and steno-occlusive vascular disease. Meeting demands for oxygen delivery requires compensatory features of brain perfusion. The cerebral vasculature’s regulatory function and reserves can be assessed by observing the flow response to a vasoactive stimulus. In a traditional approach we measured voxel-wise change in Blood Oxygen-Level Dependent (BOLD) MRI signal as a surrogate of cerebral blood flow (CBF) in response to a linear progressive ramping of end-tidal partial pressure of carbon dioxide (PETCO2). Cerebrovascular reactivity (CVR) was defined as ΔBOLD/ΔPETCO2. We used a computer model to fit a virtual sigmoid resistance curve to the progressive CBF response to the stimulus, enabling the calculation of resistance parameters: amplitude, midpoint, range response, resistance sensitivity and vasodilatory reserve. The quality of the resistance sigmoid fit was expressed as the r2 of the fit. We tested 35 patients with SCD, as well as 24 healthy subjects to provide an indication of the normal ranges of the resistance parameters. We found that gray matter CVR and resistance amplitude, range, reserve, and sensitivity are reduced in patients with SCD compared to healthy controls, while resistance midpoint was increased. This study is the first to document resistance measures in adult patients with SCD. It is also the first to score these vascular resistance measures in comparison to the normal range. We anticipate these data will complement the current understanding of the cerebral vascular pathophysiology of SCD, identify paths for therapeutic interventions, and provide biomarkers for monitoring the progress of the disease.
Collapse
Affiliation(s)
- Ece Su Sayin
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Department of Anaesthesia and Pain Management, University Health Network, Toronto, ON, Canada
| | - Olivia Sobczyk
- Department of Anaesthesia and Pain Management, University Health Network, Toronto, ON, Canada
- Joint Department of Medical Imaging and the Functional Neuroimaging Laboratory, University Health Network, Toronto, ON, Canada
| | - Julien Poublanc
- Joint Department of Medical Imaging and the Functional Neuroimaging Laboratory, University Health Network, Toronto, ON, Canada
| | - David J. Mikulis
- Joint Department of Medical Imaging and the Functional Neuroimaging Laboratory, University Health Network, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Joseph A. Fisher
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Department of Anaesthesia and Pain Management, University Health Network, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Kevin H. M. Kuo
- Division of Hematology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - James Duffin
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Department of Anaesthesia and Pain Management, University Health Network, Toronto, ON, Canada
- *Correspondence: James Duffin,
| |
Collapse
|
23
|
Chen X, Zhang J, Wu Y, Tucker R, Baird GL, Domonoske R, Barrios-Anderson A, Lim YP, Bath K, Walsh EG, Stonestreet BS. Inter-alpha Inhibitor Proteins Ameliorate Brain Injury and Improve Behavioral Outcomes in a Sex-Dependent Manner After Exposure to Neonatal Hypoxia Ischemia in Newborn and Young Adult Rats. Neurotherapeutics 2022; 19:528-549. [PMID: 35290609 PMCID: PMC9226254 DOI: 10.1007/s13311-022-01217-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 12/16/2022] Open
Abstract
Hypoxic-ischemic (HI) brain injury is a major contributor to neurodevelopmental morbidities. Inter-alpha inhibitor proteins (IAIPs) have neuroprotective effects on HI-related brain injury in neonatal rats. However, the effects of treatment with IAIPs on sequential behavioral, MRI, and histopathological abnormalities in the young adult brain after treatment with IAIPs in neonates remain to be determined. The objective of this study was to examine the neuroprotective effects of IAIPs at different neurodevelopmental stages from newborn to young adults after exposure of neonates to HI injury. IAIPs were given as 11-sequential 30-mg/kg doses to postnatal (P) day 7-21 rats after right common carotid artery ligation and exposure to 90 min of 8% oxygen. The resulting brain edema and injury were examined by T2-weighted magnetic resonance imaging (MRI) and cresyl violet staining, respectively. The mean T2 values of the ipsilateral hemisphere from MRI slices 6 to 10 were reduced in IAIP-treated HI males + females on P8, P9, and P10 and females on P8, P9, P10, and P14. IAIP treatment reduced hemispheric volume atrophy by 44.5 ± 29.7% in adult male + female P42 rats and improved general locomotor abilities measured by the righting reflex over time at P7.5, P8, and P9 in males + females and males and muscle strength/endurance measured by wire hang on P16 in males + females and females. IAIPs provided beneficial effects during the learning phase of the Morris water maze with females exhibiting beneficial effects. IAIPs confer neuroprotection from HI-related brain injury in neonates and even in adult rats and beneficial MRI and behavioral benefits in a sex-dependent manner.
Collapse
Affiliation(s)
- Xiaodi Chen
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Jiyong Zhang
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Yuqi Wu
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Richard Tucker
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Grayson L Baird
- Department of Diagnostic Imaging, Biostatistics Core Lifespan Hospital System, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Rose Domonoske
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Adriel Barrios-Anderson
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Yow-Pin Lim
- ProThera Biologics, Inc, Providence, RI, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Kevin Bath
- Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Irving Medical College, New York, NY, USA
| | - Edward G Walsh
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Barbara S Stonestreet
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA.
| |
Collapse
|
24
|
Brégère C, Schwendele B, Radanovic B, Guzman R. Microglia and Stem-Cell Mediated Neuroprotection after Neonatal Hypoxia-Ischemia. Stem Cell Rev Rep 2022; 18:474-522. [PMID: 34382141 PMCID: PMC8930888 DOI: 10.1007/s12015-021-10213-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2021] [Indexed: 12/14/2022]
Abstract
Neonatal hypoxia-ischemia encephalopathy (HIE) refers to a brain injury in term infants that can lead to death or lifelong neurological deficits such as cerebral palsy (CP). The pathogenesis of this disease involves multiple cellular and molecular events, notably a neuroinflammatory response driven partly by microglia, the brain resident macrophages. Treatment options are currently very limited, but stem cell (SC) therapy holds promise, as beneficial outcomes are reported in animal studies and to a lesser degree in human trials. Among putative mechanisms of action, immunomodulation is considered a major contributor to SC associated benefits. The goal of this review is to examine whether microglia is a cellular target of SC-mediated immunomodulation and whether the recruitment of microglia is linked to brain repair. We will first provide an overview on microglial activation in the rodent model of neonatal HI, and highlight its sensitivity to developmental age. Two complementary questions are then addressed: (i) do immune-related treatments impact microglia and provide neuroprotection, (ii) does stem cell treatment modulates microglia? Finally, the immune-related findings in patients enrolled in SC based clinical trials are discussed. Our review points to an impact of SCs on the microglial phenotype, but heterogeneity in experimental designs and methodological limitations hamper our understanding of a potential contribution of microglia to SC associated benefits. Thorough analyses of the microglial phenotype are warranted to better address the relevance of the neuroimmune crosstalk in brain repair and improve or advance the development of SC protocols in humans.
Collapse
Affiliation(s)
- Catherine Brégère
- Department of Biomedicine and Department of Neurosurgery, Faculty of Medicine, University Hospital Basel, Basel, Switzerland
| | - Bernd Schwendele
- Department of Biomedicine and Department of Neurosurgery, Faculty of Medicine, University Hospital Basel, Basel, Switzerland
| | - Boris Radanovic
- Department of Biomedicine and Department of Neurosurgery, Faculty of Medicine, University Hospital Basel, Basel, Switzerland
| | - Raphael Guzman
- Department of Biomedicine and Department of Neurosurgery, Faculty of Medicine, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
25
|
Nakanishi K, Higashi K, Toida T, Asai M. Characterization of chondroitin sulfate in stem cells derived from umbilical cord blood in rats. PLoS One 2022; 17:e0262854. [PMID: 35077481 PMCID: PMC8789104 DOI: 10.1371/journal.pone.0262854] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 01/06/2022] [Indexed: 11/26/2022] Open
Abstract
Chondroitin sulfate (CS) and its isomeric variant, dermatan sulfate (DS), are complex glycosaminoglycans (GAGs) which are ubiquitous components of the extracellular matrix in various tissues including the brain. CS and/or DS are known to bind to a variety of growth factors and regulate many cellular events such as proliferation and differentiation. Although the biological activities of CS and/or DS towards neural stem/progenitor cells (NSPCs) have been well investigated, the CS and/or DS of hematopoietic stem cells (HSCs) have not been fully characterized. Here, we analyzed GAGs on mononuclear cells of rat umbilical cord blood cells (UCB-MNCs). CS was detected in vascular intima and media of rat umbilical cord at embryonic day 19 (E19) by immunohistochemistry. The stem-cell-enriched-UCBCs (SCE-UCBCs), which were expanded from rat UCB-MNCs, expressed CS. CS chains are composed of repeating disaccharide units, which are classified into several types such as O-, A-, B-, C-, D-, and E-unit according to the number and positions of sulfation. A disaccharide composition analysis revealed that CS and/or DS were abundant in rat UCB-MNCs as well as in their expanded SCE-UCBCs, while the amount of heparan sulfate (HS) was less. The degree of sulfation of CS/DS was relatively low and the major component in UCB-MNCs and SCE-UCBCs was the A-unit. A colony-forming cell assay revealed that the percentage of colony-forming cells decreased in culture with CS degradation enzyme. The CS and/or DS of UCBCs may be involved in biological activities such as stem cell proliferation and/or differentiation.
Collapse
Affiliation(s)
- Keiko Nakanishi
- Department of Disease Model, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai, Aichi, Japan
- Department of Pediatrics, Central Hospital, Aichi Developmental Disability Center, Kasugai, Aichi, Japan
- * E-mail:
| | - Kyohei Higashi
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Toshihiko Toida
- Center for Preventive Medical Sciences, Chiba University, Chuo-ku, Chiba, Japan
| | - Masato Asai
- Department of Disease Model, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai, Aichi, Japan
| |
Collapse
|
26
|
Gamage TKJB, Fraser M. The Role of Extracellular Vesicles in the Developing Brain: Current Perspective and Promising Source of Biomarkers and Therapy for Perinatal Brain Injury. Front Neurosci 2021; 15:744840. [PMID: 34630028 PMCID: PMC8498217 DOI: 10.3389/fnins.2021.744840] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
This comprehensive review focuses on our current understanding of the proposed physiological and pathological functions of extracellular vesicles (EVs) in the developing brain. Furthermore, since EVs have attracted great interest as potential novel cell-free therapeutics, we discuss advances in the knowledge of stem cell- and astrocyte-derived EVs in relation to their potential for protection and repair following perinatal brain injury. This review identified 13 peer-reviewed studies evaluating the efficacy of EVs in animal models of perinatal brain injury; 12/13 utilized mesenchymal stem cell-derived EVs (MSC-EVs) and 1/13 utilized astrocyte-derived EVs. Animal model, method of EV isolation and size, route, timing, and dose administered varied between studies. Notwithstanding, EV treatment either improved and/or preserved perinatal brain structures both macroscopically and microscopically. Additionally, EV treatment modulated inflammatory responses and improved brain function. Collectively this suggests EVs can ameliorate, or repair damage associated with perinatal brain injury. These findings warrant further investigation to identify the optimal cell numbers, source, and dosage regimens of EVs, including long-term effects on functional outcomes.
Collapse
|
27
|
Umbilical cord blood therapy modulates neonatal hypoxic ischemic brain injury in both females and males. Sci Rep 2021; 11:15788. [PMID: 34349144 PMCID: PMC8338979 DOI: 10.1038/s41598-021-95035-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
Preclinical and clinical studies have shown that sex is a significant risk factor for perinatal morbidity and mortality, with males being more susceptible to neonatal hypoxic ischemic (HI) brain injury. No study has investigated sexual dimorphism in the efficacy of umbilical cord blood (UCB) cell therapy. HI injury was induced in postnatal day 10 (PND10) rat pups using the Rice-Vannucci method of carotid artery ligation. Pups received 3 doses of UCB cells (PND11, 13, 20) and underwent behavioural testing. On PND50, brains were collected for immunohistochemical analysis. Behavioural and neuropathological outcomes were assessed for sex differences. HI brain injury resulted in a significant decrease in brain weight and increase in tissue loss in females and males. Females and males also exhibited significant cell death, region-specific neuron loss and long-term behavioural deficits. Females had significantly smaller brains overall compared to males and males had significantly reduced neuron numbers in the cortex compared to females. UCB administration improved multiple aspects of neuropathology and functional outcomes in males and females. Females and males both exhibited injury following HI. This is the first preclinical evidence that UCB is an appropriate treatment for neonatal brain injury in both female and male neonates.
Collapse
|
28
|
Robertson NJ, Meehan C, Martinello KA, Avdic-Belltheus A, Boggini T, Mutshiya T, Lingam I, Yang Q, Sokolska M, Charalambous X, Bainbridge A, Hristova M, Kramer BW, Golay X, Weil B, Lowdell MW. Human umbilical cord mesenchymal stromal cells as an adjunct therapy with therapeutic hypothermia in a piglet model of perinatal asphyxia. Cytotherapy 2021; 23:521-535. [PMID: 33262073 PMCID: PMC8139415 DOI: 10.1016/j.jcyt.2020.10.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 10/12/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND With therapeutic hypothermia (HT) for neonatal encephalopathy, disability rates are reduced, but not all babies benefit. Pre-clinical rodent studies suggest mesenchymal stromal cells (MSCs) augment HT protection. AIMS The authors studied the efficacy of intravenous (IV) or intranasal (IN) human umbilical cord-derived MSCs (huMSCs) as adjunct therapy to HT in a piglet model. METHODS A total of 17 newborn piglets underwent transient cerebral hypoxia-ischemia (HI) and were then randomized to (i) HT at 33.5°C 1-13 h after HI (n = 7), (ii) HT+IV huMSCs (30 × 106 cells) at 24 h and 48 h after HI (n = 5) or (iii) HT+IN huMSCs (30 × 106 cells) at 24 h and 48 h after HI (n = 5). Phosphorus-31 and hydrogen-1 magnetic resonance spectroscopy (MRS) was performed at 30 h and 72 h and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells and oligodendrocytes quantified. In two further piglets, 30 × 106 IN PKH-labeled huMSCs were administered. RESULTS HI severity was similar between groups. Amplitude-integrated electroencephalogram (aEEG) recovery was more rapid for HT+IN huMSCs compared with HT from 25 h to 42 h and 49 h to 54 h (P ≤ 0.05). MRS phosphocreatine/inorganic phosphate was higher on day 2 in HT+IN huMSCs than HT (P = 0.035). Comparing HT+IN huMSCs with HT and HT+IV huMSCs, there were increased OLIG2 counts in hippocampus (P = 0.011 and 0.018, respectively), internal capsule (P = 0.013 and 0.037, respectively) and periventricular white matter (P = 0.15 for IN versus IV huMSCs). Reduced TUNEL-positive cells were seen in internal capsule with HT+IN huMSCs versus HT (P = 0.05). PKH-labeled huMSCs were detected in the brain 12 h after IN administration. CONCLUSIONS After global HI, compared with HT alone, the authors saw beneficial effects of HT+IN huMSCs administered at 24 h and 48 h (30 × 106 cells/kg total dose) based on more rapid aEEG recovery, improved 31P MRS brain energy metabolism and increased oligodendrocyte survival at 72 h.
Collapse
Affiliation(s)
| | | | | | | | - Tiziana Boggini
- Institute for Women's Health, University College London, London, UK
| | - Tatenda Mutshiya
- Institute for Women's Health, University College London, London, UK
| | - Ingran Lingam
- Institute for Women's Health, University College London, London, UK
| | - Qin Yang
- Institute for Women's Health, University College London, London, UK
| | | | | | - Alan Bainbridge
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Mariya Hristova
- Institute for Women's Health, University College London, London, UK
| | - Boris W Kramer
- Department of Pediatrics, University of Maastricht, Maastricht, the Netherlands
| | - Xavier Golay
- Institute for Women's Health, University College London, London, UK
| | - Ben Weil
- Royal Free London NHS Foundation Trust, London, UK
| | - Mark W Lowdell
- Institute for Women's Health, University College London, London, UK; Royal Free London NHS Foundation Trust, London, UK
| |
Collapse
|
29
|
Sato Y, Tsuji M. Diverse actions of cord blood cell therapy for hypoxic-ischemic encephalopathy. Pediatr Int 2021; 63:497-503. [PMID: 33453136 PMCID: PMC8252712 DOI: 10.1111/ped.14604] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/25/2020] [Accepted: 01/12/2021] [Indexed: 12/18/2022]
Abstract
Perinatal hypoxic-ischemic encephalopathy (HIE) is a major cause of neonatal death and permanent neurological deficits. However, effective treatments have not yet been established, except therapeutic hypothermia, which is not effective for severe HIE; therefore, developing a novel therapy for HIE is of the utmost importance. Stem cell therapy has recently been identified as a novel therapy for HIE. Among the various stem cell sources, ethical hurdles can be avoided by using stem cells that originate from non-embryonic or non-neural tissues, such as umbilical cord blood cells (UCBCs), which are readily available and can be exploited for autologous transplantations. Human UCBs are a rich source of stem and progenitor cells. Many recent studies have reported the treatment effect of UCBCs. Additionally, phase I clinical trials have already been conducted, showing this therapy's safety and feasibility. One advantage of stem cell therapies, including UCBC administration, is that they exert treatment effects through multifaceted mechanisms. According to the findings of several publications, replacement of lost cells, namely, engraftment and differentiation into neuronal cells, is not likely to be the main mechanism. However, the association between UCBCs and various mechanism of action, such as neurogenesis, angiogenesis, and anti-inflammation, has been suggested in many studies, and most mechanisms are due to growth factors secreted from UCBCs. These diverse actions of UCBC treatment are expected to exert a substantial effect on HIE, which has a complex injury mechanism.
Collapse
Affiliation(s)
- Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Masahiro Tsuji
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| |
Collapse
|
30
|
Penny TR, Pham Y, Sutherland AE, Smith MJ, Lee J, Jenkin G, Fahey MC, Miller SL, McDonald CA. Optimization of behavioral testing in a long-term rat model of hypoxic ischemic brain injury. Behav Brain Res 2021; 409:113322. [PMID: 33901432 DOI: 10.1016/j.bbr.2021.113322] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/02/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Hypoxic ischemic (HI) brain injury is a significant cause of childhood neurological deficits. Preclinical rodent models are often used to study these deficits; however, no preclinical study has determined which behavioral tests are most appropriate for long-term follow up after neonatal HI. METHODS HI brain injury was induced in postnatal day (PND) 10 rat pups using the Rice-Vannucci method of unilateral carotid artery ligation. Rats underwent long-term behavioral testing to assess motor and cognitive outcomes between PND11-50. Behavioral scores were transformed into Z-scores and combined to create composite behavioral scores. RESULTS HI rats showed a significant deficit in three out of eight behavioral tests: negative geotaxis analysis, the cylinder test and the novel object recognition test. These individual test outcomes were transformed into Z-scores and combined to create a composite Z-score. This composite z-score showed that HI rats had a significantly increased behavioral burden over the course of the experiment. CONCLUSION In this study we have identified tests that highlight specific cognitive and motor deficits in a rat model of neonatal HI. Due to the high variability in this model of neonatal HI brain injury, significant impairment is not always observed in individual behavioral tests, but by combining outcomes from these individual tests, long-term behavioral burden can be measured.
Collapse
Affiliation(s)
- Tayla R Penny
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Yen Pham
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Amy E Sutherland
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Madeleine J Smith
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Joohyung Lee
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Michael C Fahey
- Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Courtney A McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
| |
Collapse
|
31
|
Stem Cell Therapy for Neonatal Hypoxic-Ischemic Encephalopathy: A Systematic Review of Preclinical Studies. Int J Mol Sci 2021; 22:ijms22063142. [PMID: 33808671 PMCID: PMC8003344 DOI: 10.3390/ijms22063142] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is an important cause of mortality and morbidity in the perinatal period. This condition results from a period of ischemia and hypoxia to the brain of neonates, leading to several disorders that profoundly affect the daily life of patients and their families. Currently, therapeutic hypothermia (TH) is the standard of care in developing countries; however, TH is not always effective, especially in severe cases of HIE. Addressing this concern, several preclinical studies assessed the potential of stem cell therapy (SCT) for HIE. With this systematic review, we gathered information included in 58 preclinical studies from the last decade, focusing on the ones using stem cells isolated from the umbilical cord blood, umbilical cord tissue, placenta, and bone marrow. Outstandingly, about 80% of these studies reported a significant improvement of cognitive and/or sensorimotor function, as well as decreased brain damage. These results show the potential of SCT for HIE and the possibility of this therapy, in combination with TH, becoming the next therapeutic approach for HIE. Nonetheless, few preclinical studies assessed the combination of TH and SCT for HIE, and the existent studies show some contradictory results, revealing the need to further explore this line of research.
Collapse
|
32
|
Segler A, Braun T, Fischer HS, Dukatz R, Weiss CR, Schwickert A, Jäger C, Bührer C, Henrich W. Feasibility of Umbilical Cord Blood Collection in Neonates at Risk of Brain Damage-A Step Toward Autologous Cell Therapy for a High-risk Population. Cell Transplant 2021; 30:963689721992065. [PMID: 33631961 PMCID: PMC7917411 DOI: 10.1177/0963689721992065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Evidence for umbilical cord blood (UCB) cell therapies as a potential intervention for neurological diseases is emerging. To date, most existing trials worked with allogenic cells, as the collection of autologous UCB from high-risk patients is challenging. In obstetric emergencies the collection cannot be planned. In preterm infants, late cord clamping and anatomic conditions may reduce the availability. The aim of the present study was to assess the feasibility of UCB collection in neonates at increased risk of brain damage. Infants from four high-risk groups were included: newborns with perinatal hypoxemia, gestational age (GA) ≤30 + 0 weeks and/or birthweight <1,500 g, intrauterine growth restriction (IUGR), or monochorionic twins with twin-to-twin transfusion syndrome (TTTS). Feasibility of collection, quantity and quality of obtained UCB [total nucleated cell count (TNC), volume, sterility, and cell viability], and neonatal outcome were assessed. UCB collection was successful in 141 of 177 enrolled patients (hypoxemia n = 10; GA ≤30 + 0 weeks n = 54; IUGR n = 71; TTTS n = 6). Twenty-six cases were missed. The amount of missed cases per month declined over the time. Volume of collected UCB ranged widely (median: 24.5 ml, range: 5.0–102 ml) and contained a median of 0.77 × 108 TNC (range: 0.01–13.0 × 108). TNC and UCB volume correlated significantly with GA. A total of 10.7% (19/177) of included neonates developed brain lesions. To conclude, collection of UCB in neonates at high risk of brain damage is feasible with a multidisciplinary approach and intensive training. High prevalence of brain damage makes UCB collection worthwhile. Collected autologous UCB from mature neonates harbors a sufficient cell count for potential therapy. However, quality and quantity of obtained UCB are critical for potential therapy in preterm infants. Therefore, for extremely preterm infants alternative cell sources such as UCB tissue should be investigated for autologous treatment options because of the low yield of UCB.
Collapse
Affiliation(s)
- Angela Segler
- Department of Obstetrics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Thorsten Braun
- Department of Obstetrics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of "Experimental Obstetrics" and Study group "Perinatal Programming", Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Hendrik Stefan Fischer
- Department of Neonatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ricarda Dukatz
- Department of Obstetrics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Claire-Rachel Weiss
- Department of Obstetrics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alexander Schwickert
- Department of Obstetrics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Carsten Jäger
- Department of Surgery, Klinikum rechts der Isar, Technische Universitaüt Muünchen, Munich, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Wolfgang Henrich
- Department of Obstetrics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
33
|
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
|
34
|
Shintaku H. Prevention and treatment of cerebral palsy with cord blood stem cells and cord-derived mesenchymal stem cells. Neural Regen Res 2021; 16:672-673. [PMID: 33063719 PMCID: PMC8067942 DOI: 10.4103/1673-5374.293139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Haruo Shintaku
- Donated Course "Disability medicine and Regenerative medicine", Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
35
|
Penny TR, Pham Y, Sutherland AE, Mihelakis JG, Lee J, Jenkin G, Fahey MC, Miller SL, McDonald CA. Multiple doses of umbilical cord blood cells improve long-term brain injury in the neonatal rat. Brain Res 2020; 1746:147001. [PMID: 32585139 DOI: 10.1016/j.brainres.2020.147001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/11/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hypoxic ischemic (HI) insults during pregnancy and birth can result in neurodevelopmental disorders, such as cerebral palsy. We have previously shown that a single dose of umbilical cord blood (UCB) cells is effective at reducing short-term neuroinflammation and improves short and long-term behavioural outcomes in rat pups. A single dose of UCB was not able to modulate long-term neuroinflammation or brain tissue loss. In this study we examined whether multiple doses of UCB can modulate neuroinflammation, decrease cerebral tissue damage and improve behavioural outcomes when followed up long-term. METHODS HI injury was induced in postnatal day 10 (PND10) rat pups using the Rice-Vannucci method of carotid artery ligation. Pups received either 1 dose (PND11), or 3 doses (PND11, 13, 20) of UCB cells. Rats were followed with behavioural testing, to assess both motor and cognitive outcomes. On PND50, brains were collected for analysis. RESULTS HI brain injury in rat pups caused significant behavioural deficits. These deficits were significantly improved by multiple doses of UCB. HI injury resulted in a significant decrease in brain weight and left hemisphere tissue, which was improved by multiple doses of UCB. HI resulted in increased cerebral apoptosis, loss of neurons and upregulation of activated microglia. Multiple doses of UCB modulated these neuropathologies. A single dose of UCB at PND11 did not improve behavioural or neuropathological outcomes. CONCLUSIONS Treatment with repeated doses of UCB is more effective than a single dose for reducing tissue damage, improving brain pathology and restoring behavioural deficits following perinatal brain injury.
Collapse
Affiliation(s)
- Tayla R Penny
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Yen Pham
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Amy E Sutherland
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Jamie G Mihelakis
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Joohyung Lee
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Michael C Fahey
- Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Courtney A McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
| |
Collapse
|
36
|
TNF-α Pretreatment Improves the Survival and Function of Transplanted Human Neural Progenitor Cells Following Hypoxic-Ischemic Brain Injury. Cells 2020; 9:cells9051195. [PMID: 32403417 PMCID: PMC7291333 DOI: 10.3390/cells9051195] [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: 03/09/2020] [Revised: 04/28/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023] Open
Abstract
Neural progenitor cells (NPCs) therapy offers great promise in hypoxic-ischemic (HI) brain injury. However, the poor survival of implanted NPCs in the HI host environment limits their therapeutic effects. Tumor necrosis factor-alpha (TNF-α) is a pleiotropic cytokine that is induced in response to a variety of pathological processes including inflammation and immunity. On the other hand, TNF-α has protective effects on cell apoptosis and death and affects the differentiation, proliferation, and survival of neural stem/progenitor cells in the brain. The present study investigated whether TNF-α pretreatment on human NPCs (hNPCs) enhances the effectiveness of cell transplantation therapy under ischemic brain. Fetal brain tissue-derived hNPCs were pretreated with TNF-α before being used in vitro experiments or transplantation. TNF-α significantly increased expression of cIAP2, and the use of short hairpin RNA-mediated knockdown of cIAP2 demonstrated that cIAP2 protected hNPCs against HI-induced cytotoxicity. In addition, pretreatment of hNPCs with TNF-α mediated neuroprotection by altering microglia polarization via increased expression of CX3CL1 and by enhancing expression of neurotrophic factors. Furthermore, transplantation of TNF-α-treated hNPCs reduced infarct volume and improved neurological functions in comparison with non-pretreated hNPCs or vehicle. These findings show that TNF-α pretreatment, which protects hNPCs from HI-injured brain-induced apoptosis and increases neuroprotection, is a simple and safe approach to improve the survival of transplanted hNPCs and the therapeutic efficacy of hNPCs in HI brain injury.
Collapse
|
37
|
Greco P, Nencini G, Piva I, Scioscia M, Volta CA, Spadaro S, Neri M, Bonaccorsi G, Greco F, Cocco I, Sorrentino F, D'Antonio F, Nappi L. Pathophysiology of hypoxic-ischemic encephalopathy: a review of the past and a view on the future. Acta Neurol Belg 2020; 120:277-288. [PMID: 32112349 DOI: 10.1007/s13760-020-01308-3] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/13/2020] [Indexed: 12/14/2022]
Abstract
Hypoxic-ischemic encephalopathy, also referred as HIE, is a type of brain injury or damage that is caused by a lack of oxygen to the brain during neonatal period. The incidence is approximately 1.5 cases per 1000 live births in developed countries. In low and middle-income countries, the incidence is much higher (10‒20 per 1000 live births). The treatment for neonatal HIE is hypothermia that is only partially effective (not more than 50% of the neonates treated achieve an improved outcome). HIE pathophysiology involves oxidative stress, mitochondrial energy production failure, glutaminergic excitotoxicity, and apoptosis. So, in the last years, many studies have focused on peptides that act somewhere in the pathway activated by severe anoxic injury leading to HIE. This review describes the pathophysiology of perinatal HIE and the mechanisms that could be the target of innovative HIE treatments.
Collapse
Affiliation(s)
- P Greco
- Department of Morphology, Surgery and Experimental Medicine, Institute of Obstetrics and Gynaecology, University of Ferrara, 44121, Ferrara, Italy
| | - G Nencini
- Department of Morphology, Surgery and Experimental Medicine, Institute of Obstetrics and Gynaecology, University of Ferrara, 44121, Ferrara, Italy
| | - I Piva
- Department of Women Health, Infancy and Adolescence, AUSL Ravenna, 48121, Ravenna, Italy
| | - M Scioscia
- Department of Obstetrics and Gynaecology, Policlinico Hospital of Abano Terme, Padua, Italy
| | - C A Volta
- Section of Anesthesia and Intensive Care, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121, Ferrara, Italy
| | - S Spadaro
- Section of Anesthesia and Intensive Care, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121, Ferrara, Italy
| | - M Neri
- Section of Forensic Medicine, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121, Ferrara, Italy
| | - G Bonaccorsi
- Department of Morphology, Surgery and Experimental Medicine, Institute of Obstetrics and Gynaecology, University of Ferrara, 44121, Ferrara, Italy
| | - F Greco
- Department of Medical and Surgical Sciences, Institute of Obstetrics and Gynaecology, University of Foggia, 71121, Foggia, Italy
| | - I Cocco
- Department of Medical and Surgical Sciences, Institute of Obstetrics and Gynaecology, University of Foggia, 71121, Foggia, Italy
| | - F Sorrentino
- Department of Medical and Surgical Sciences, Institute of Obstetrics and Gynaecology, University of Foggia, 71121, Foggia, Italy.
| | - F D'Antonio
- Department of Medical and Surgical Sciences, Institute of Obstetrics and Gynaecology, University of Foggia, 71121, Foggia, Italy
| | - L Nappi
- Department of Medical and Surgical Sciences, Institute of Obstetrics and Gynaecology, University of Foggia, 71121, Foggia, Italy
| |
Collapse
|
38
|
Tsuji M, Sawada M, Watabe S, Sano H, Kanai M, Tanaka E, Ohnishi S, Sato Y, Sobajima H, Hamazaki T, Mori R, Oka A, Ichiba H, Hayakawa M, Kusuda S, Tamura M, Nabetani M, Shintaku H. Autologous cord blood cell therapy for neonatal hypoxic-ischaemic encephalopathy: a pilot study for feasibility and safety. Sci Rep 2020; 10:4603. [PMID: 32165664 PMCID: PMC7067794 DOI: 10.1038/s41598-020-61311-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/03/2020] [Indexed: 02/02/2023] Open
Abstract
Neonatal hypoxic-ischaemic encephalopathy (HIE) is a serious condition; many survivors develop neurological impairments, including cerebral palsy and intellectual disability. Preclinical studies show that the systemic administration of umbilical cord blood cells (UCBCs) is beneficial for neonatal HIE. We conducted a single-arm clinical study to examine the feasibility and safety of intravenous infusion of autologous UCBCs for newborns with HIE. When a neonate was born with severe asphyxia, the UCB was collected, volume-reduced, and divided into three doses. The processed UCB was infused at 12–24, 36–48, and 60–72 hours after the birth. The designed enrolment was six newborns. All six newborns received UCBC therapy strictly adhering to the study protocol together with therapeutic hypothermia. The physiological parameters and peripheral blood parameters did not change much between pre- and postinfusion. There were no serious adverse events that might be related to cell therapy. At 30 days of age, the six infants survived without circulatory or respiratory support. At 18 months of age, neurofunctional development was normal without any impairment in four infants and delayed with cerebral palsy in two infants. This pilot study shows that autologous UCBC therapy is feasible and safe.
Collapse
Affiliation(s)
- Masahiro Tsuji
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, 605-8501, Japan.,Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Suita, 565-8565, Japan
| | - Mariko Sawada
- Department of Pediatrics, Kurashiki Central Hospital, Kurashiki, 710-8602, Japan
| | - Shinichi Watabe
- Department of Pediatrics, Kurashiki Central Hospital, Kurashiki, 710-8602, Japan
| | - Hiroyuki Sano
- Department of Pediatrics, Yodogawa Christian Hospital, Osaka, 533-0024, Japan
| | - Masayo Kanai
- Division of Neonatology, Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Kawagoe, 350-8850, Japan
| | - Emi Tanaka
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, 545-8585, Japan
| | - Satoshi Ohnishi
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, 545-8585, Japan
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, 466-8560, Japan
| | - Hisanori Sobajima
- Division of Neonatology, Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Kawagoe, 350-8850, Japan
| | - Takashi Hamazaki
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, 545-8585, Japan
| | - Rintaro Mori
- Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Akira Oka
- Department of Pediatrics, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Hiroyuki Ichiba
- Department of Neonatology, Osaka City General Hospital, Osaka, 534-0021, Japan
| | - Masahiro Hayakawa
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, 466-8560, Japan
| | - Satoshi Kusuda
- Department of Pediatrics, Kyorin University, Mitaka, 181-8611, Japan
| | - Masanori Tamura
- Division of Neonatology, Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Kawagoe, 350-8850, Japan
| | - Makoto Nabetani
- Department of Pediatrics, Yodogawa Christian Hospital, Osaka, 533-0024, Japan.
| | - Haruo Shintaku
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, 545-8585, Japan.
| |
Collapse
|
39
|
Crompton K, Novak I, Fahey M, Badawi N, Wallace E, Lee K, Mechinaud-Heloury F, Colditz PB, Elwood N, Edwards P, Reddihough D. Single group multisite safety trial of sibling cord blood cell infusion to children with cerebral palsy: study protocol and rationale. BMJ Open 2020; 10:e034974. [PMID: 32152173 PMCID: PMC7064081 DOI: 10.1136/bmjopen-2019-034974] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/10/2019] [Accepted: 02/04/2020] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION Cerebral palsy (CP) is the most common physical disability of childhood but has no cure. Stem cells have the potential to improve brain injury and are proposed as a therapy for CP. However, many questions remain unanswered about the most appropriate cell type, timing of infusions, dose required and associated risks. Therefore, human safety and efficacy trials are necessary to progress knowledge in the field. METHODS AND ANALYSIS This is a single group study with sample size n=12 to investigate safety of single-dose intravenous 12/12 human leucocyte antigen-matched sibling cord blood cell infusion to children with CP aged 1-16 years without immune suppression. The study is similar to a 3+3 design, where the first two groups of participants have severe CP, and the final six participants include children with all motor severities. Children will be monitored for adverse events and the duration that donor cells are detected. Assessments at baseline, 3 and 12 months will investigate safety and preliminary evidence of change in gross motor, fine motor, cognitive and quality of life outcomes. ETHICS AND DISSEMINATION Full approval was obtained from The Royal Children's Hospital Human Research Ethics Committee, and a clinical trial notification was accepted by Australia's Therapeutic Goods Administration. Participant guardian informed consent will be obtained before any study procedures. The main results of this study will be submitted for publication in a peer-reviewed journal. TRIAL REGISTRATION NUMBER ACTRN12616000403437, NCT03087110.
Collapse
Affiliation(s)
- Kylie Crompton
- Neurodisability and Rehabilitation, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Neurodevelopment and Disability, The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
- Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Iona Novak
- Cerebral Palsy Alliance Research Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Michael Fahey
- Paediatric Neurology, Monash Health, Clayton, Victoria, Australia
- Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Nadia Badawi
- Cerebral Palsy Alliance Research Institute, The University of Sydney, Sydney, New South Wales, Australia
- Grace Centre for Newborn Care, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Euan Wallace
- Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Katherine Lee
- Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | | | - Paul B Colditz
- Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Ngaire Elwood
- Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Cell Biology, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Priya Edwards
- Queensland Paediatric Rehabilitation Service, Children's Health Queensland Hospital and Health Service, Herston, Queensland, Australia
- Queensland Cerebral Palsy and Rehabilitation Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Dinah Reddihough
- Neurodisability and Rehabilitation, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Neurodevelopment and Disability, The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
- Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
40
|
Arteaga Cabeza O, Mikrogeorgiou A, Kannan S, Ferriero DM. Advanced nanotherapies to promote neuroregeneration in the injured newborn brain. Adv Drug Deliv Rev 2019; 148:19-37. [PMID: 31678359 DOI: 10.1016/j.addr.2019.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/19/2019] [Accepted: 10/23/2019] [Indexed: 12/16/2022]
Abstract
Neonatal brain injury affects thousands of babies each year and may lead to long-term and permanent physical and neurological problems. Currently, therapeutic hypothermia is standard clinical care for term newborns with moderate to severe neonatal encephalopathy. Nevertheless, it is not completely protective, and additional strategies to restore and promote regeneration are urgently needed. One way to ensure recovery following injury to the immature brain is to augment endogenous regenerative pathways. However, novel strategies such as stem cell therapy, gene therapies and nanotechnology have not been adequately explored in this unique age group. In this perspective review, we describe current efforts that promote neuroprotection and potential targets that are unique to the developing brain, which can be leveraged to facilitate neuroregeneration.
Collapse
|
41
|
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
|
42
|
Effects of human umbilical cord blood CD34 + cell transplantation in neonatal hypoxic-ischemia rat model. Brain Dev 2019; 41:173-181. [PMID: 30177297 DOI: 10.1016/j.braindev.2018.08.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 08/16/2018] [Indexed: 12/18/2022]
Abstract
Perinatal brain injury can cause death in the neonatal period and lifelong neurodevelopmental deficits. Stem cell transplantation had been proved to be effective approach to ameliorate neurological deficits after brain damage. In this study we examine the effect of human umbilical cord blood CD34+ cells on model of neonatal rat hypoxic-ischemic brain damage and compared the neuroprotection of transplantation of CD34+ cells to mononuclear cells from which CD34+ cells isolated on neonatal hypoxic-ischemia rat model. Seven-day-old Sprague-Dawley rats were subjected to hypoxic-ischemic (HI) injury, CD34+ cells (1.5 × 104 cells) or mononuclear cells (1.0 × 106 cells) were transplanted into mice by tail vein on the 7 day after HI. The transplantation of CD34+ cells significantly improved motor function of rat, and reduced cerebral atrophy, inhibited the expression of glial fibrillary acidic protein (GFAP) and apoptosis-related genes: TNF-α, TNFR1, TNFR2, CD40, Fas, and decreased the activation of Nuclear factor kappa B (NF-κB) in damaged brain. CD34+ cells treatment increased the expression of DCX and lectin in ipsilateral brain. Moreover, the transplantation of CD34+ cells and MNCs which were obtained from the same amount of human umbilical cord blood had similar effects on HI. Our data demonstrated that transplantation of human umbilical cord blood CD34+ cells can ameliorate the neural functional defect and reduce apoptosis and promote nerve and vascular regeneration in rat brain after HI injury and the effects of transplantation of CD34+ cells were comparable to that of MNCs in neonatal hypoxic-ischemia rat model.
Collapse
|
43
|
Wang Q, Lv H, Lu L, Ren P, Li L. Neonatal hypoxic-ischemic encephalopathy: emerging therapeutic strategies based on pathophysiologic phases of the injury. J Matern Fetal Neonatal Med 2018; 32:3685-3692. [PMID: 29681183 DOI: 10.1080/14767058.2018.1468881] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neonatal hypoxic ischemic encephalopathy (HIE) is an important cause of neonatal death and disability. At present, there is no unified standard and specialized treatment method for neonatal HIE. In clinical practice, we have found that a gap remains between preclinical medical research and clinical application in the treatment of neonatal HIE. To promote an organic combination of preclinical research and clinical application, we propose the different phases as intervention targets, based on the pathophysiologic changes in phases I, II, and III of neonatal HIE; moreover, we suggest transformative medicine as a principle that may improve the therapeutic effect by blocking the progression of the disease to an irreversible stage. For instance, in phase I, mild hypothermia, free radical scavenger (erythropoietin, hydrogen-rich saline), excitatory amino acid receptor blocker, and neuroprotective agents should be administered to neonates with moderate/severe HIE; in phase II, following phase I treatment, anti-inflammatory agents, neuroprotective or nerve regeneration agents, and stem cell transplantation should be administered to patients; in phase III, anti-inflammatory agents, neuroprotective or nerve regeneration agents, and stem cell transplantation should be administered to patients. As soon as the patient's condition has stabilized, acupuncture, massage, and rehabilitation training should be performed. Following further study of stem cells, stem cell transplantation is expected to become the most promising therapeutic candidate for treatment of severe neonatal HIE with its sequelae.
Collapse
Affiliation(s)
- Qiuli Wang
- a Department of Neonatology , Handan Maternal and Child Health Care Hospital , Handan , PR China
| | - Hongyan Lv
- a Department of Neonatology , Handan Maternal and Child Health Care Hospital , Handan , PR China.,b Department of Neonatal Pathology , Handan Maternal and Child Health Care Hospital , Handan , PR China
| | - Lixin Lu
- c Department of Pediatrics , Handan 7th Hospital , Handan , PR China
| | - Pengshun Ren
- a Department of Neonatology , Handan Maternal and Child Health Care Hospital , Handan , PR China
| | - Lianxiang Li
- b Department of Neonatal Pathology , Handan Maternal and Child Health Care Hospital , Handan , PR China.,d Department of Neural Development and Neural Pathology , Hebei University of Engineering School of Medicine , Handan , PR China
| |
Collapse
|
44
|
Han XR, Wen X, Wang YJ, Wang S, Shen M, Zhang ZF, Fan SH, Shan Q, Wang L, Li MQ, Hu B, Sun CH, Wu DM, Lu J, Zheng YL. MicroRNA-140-5p elevates cerebral protection of dexmedetomidine against hypoxic-ischaemic brain damage via the Wnt/β-catenin signalling pathway. J Cell Mol Med 2018. [PMID: 29536658 PMCID: PMC5980153 DOI: 10.1111/jcmm.13597] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Hypoxia–ischaemia (HI) remains a major cause of foetal brain damage presented a scarcity of effective therapeutic approaches. Dexmedetomidine (DEX) and microRNA‐140‐5p (miR‐140‐5p) have been highlighted due to its potentially significant role in the treatment of cerebral ischaemia. This study was to investigate the role by which miR‐140‐5p provides cerebral protection using DEX to treat hypoxic–ischaemic brain damage (HIBD) in neonatal rats via the Wnt/β‐catenin signalling pathway. The HIBD rat models were established and allocated into various groups with different treatment plans, and eight SD rats into sham group. The learning and memory ability of the rats was assessed. Apoptosis and pathological changes in the hippocampus CA1 region and expressions of the related genes of the Wnt/β‐catenin signalling pathway as well as the genes responsible of apoptosis were detected. Compared with the sham group, the parameters of weight, length growth, weight ratio between hemispheres, the rate of reaching standard, as well as Bcl‐2 expressions, were all increased. Furthermore, observations of increased levels of cerebral infarction volume, total mortality rate, response times, total response duration, expressions of Wnt1, β‐catenin, TCF‐4, E‐cadherin, apoptosis rate of neurons, and Bax expression were elevated. Following DEX treatment, the symptoms exhibited by HIBD rats were ameliorated. miR‐140‐5p and si‐Wnt1 were noted to attenuate the progression of HIBD. Our study demonstrates that miR‐140‐5p promotes the cerebral protective effects of DEX against HIBD in neonatal rats by targeting the Wnt1 gene through via the negative regulation of the Wnt/β‐catenin signalling pathway.
Collapse
Affiliation(s)
- Xin-Rui Han
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Xin Wen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Yong-Jian Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Shan Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Min Shen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Zi-Feng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Shao-Hua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Qun Shan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Liang Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Meng-Qiu Li
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Bin Hu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Chun-Hui Sun
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Dong-Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| | - Yuan-Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu Province, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province, China
| |
Collapse
|
45
|
McDonald CA, Penny TR, Paton MCB, Sutherland AE, Nekkanti L, Yawno T, Castillo-Melendez M, Fahey MC, Jones NM, Jenkin G, Miller SL. Effects of umbilical cord blood cells, and subtypes, to reduce neuroinflammation following perinatal hypoxic-ischemic brain injury. J Neuroinflammation 2018; 15:47. [PMID: 29454374 PMCID: PMC5816393 DOI: 10.1186/s12974-018-1089-5] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 02/05/2018] [Indexed: 12/22/2022] Open
Abstract
Background It is well understood that hypoxic-ischemic (HI) brain injury during the highly vulnerable perinatal period can lead to cerebral palsy, the most prevalent cause of chronic disability in children. Recently, human clinical trials have reported safety and some efficacy following treatment of cerebral palsy using umbilical cord blood (UCB) cells. UCB is made up of many different cell types, including endothelial progenitor cells (EPCs), T regulatory cells (Tregs), and monocyte-derived suppressor cells (MDSCs). How each cell type contributes individually towards reducing neuroinflammation and/or repairing brain injury is not known. In this study, we examined whether human (h) UCB, or specific UCB cell types, could reduce peripheral and cerebral inflammation, and promote brain repair, when given early after perinatal HI brain injury. Methods HI brain injury was induced in postnatal day (PND) 7 rat pups and cells were administered intraperitoneally on PND 8. Behavioral testing was performed 7 days post injury, and then, brains and spleens were collected for analysis. Results We found in vitro that all UCB cell types, except for EPCs, were immunomodulatory. Perinatal HI brain injury induced significant infiltration of CD4+ T cells into the injured cerebral hemisphere, and this was significantly reduced by all hUCB cell types tested. Compared to HI, UCB, Tregs, and EPCs were able to reduce motor deficits, reduce CD4+ T cell infiltration into the brain, and reduce microglial activation. In addition to the beneficial effects of UCB, EPCs also significantly reduced cortical cell death, returned CD4+ T cell infiltration to sham levels, and reduced the peripheral Th1-mediated pro-inflammatory shift. Conclusion This study highlights that cells found in UCB is able to mediate neuroinflammation and is an effective neuroprotective therapy. Our study also shows that particular cells found in UCB, namely EPCs, may have an added advantage over using UCB alone. This work has the potential to progress towards tailored UCB therapies for the treatment of perinatal brain injury.
Collapse
Affiliation(s)
- Courtney A McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Victoria, 3168, Australia. .,Department of Obstetrics and Gynaecology, Monash University, Clayton, 3168, Australia.
| | - Tayla R Penny
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Victoria, 3168, Australia
| | - Madison C B Paton
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Victoria, 3168, Australia
| | - Amy E Sutherland
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Victoria, 3168, Australia
| | - Lakshmi Nekkanti
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Victoria, 3168, Australia
| | - Tamara Yawno
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Victoria, 3168, Australia
| | - Margie Castillo-Melendez
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Victoria, 3168, Australia
| | - Michael C Fahey
- Department of Paediatrics, Monash University, Clayton, 3168, Australia
| | - Nicole M Jones
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, 2052, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, 3168, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Victoria, 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, 3168, Australia
| |
Collapse
|
46
|
Qiao L, Fu J, Xue X, Shi Y, Yao L, Huang W, Li J, Zhang D, Liu N, Tong X, Du Y, Pan Y. Neuronalinjury and roles of apoptosis and autophagy in a neonatal rat model of hypoxia-ischemia-induced periventricular leukomalacia. Mol Med Rep 2018; 17:5940-5949. [PMID: 29436652 PMCID: PMC5866039 DOI: 10.3892/mmr.2018.8570] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 06/20/2017] [Indexed: 11/06/2022] Open
Abstract
As research into periventricular leukomalacia (PVL) gradually increases, concerns are emerging about long‑term neuron injury. The present study aimed to investigate neuronal injury and the relevant alterations in apoptosis and autophagy in a PVL model established previously. A rat model of hypoxia‑ischemia‑induced PVL was established. In the model group, Sprague‑Dawley (SD) rats [postnatal day 3 (P3)] were subjected to right common carotid artery ligation followed by suturing and exposed to 6‑8% oxygen for 2 h; in the control group, SD rats (P3) were subjected to right common carotid artery dissection followed by suturing, without ligation and hypoxic exposure. At 1, 3, 7 and 14 days following modeling, brain tissue samples were collected and stained with hematoxylin and eosin. Cellular apoptosis was detected by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay and the protein and mRNA expression alterations of neuronal nuclei (NeuN), caspase‑3 and Beclin 1 in the model group were detected by western blot analysis and reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) analyses. Compared with the control group, the protein and mRNA expression levels of NeuN (a marker of mature neurons) were markedly reduced, the number of positive cells was increased as detected by TUNEL, and the protein and mRNA expression levels of caspase‑3 and Beclin 1 were elevated in the model group. In the rat model of hypoxia‑ischemia‑induced PVL, oligodendrocyte injury and myelinization disorders were observed, in addition to neuron injury, a decrease in mature neurons and the co‑presence of apoptosis and autophagy. However, apoptosis and autophagy exist in different phases: Apoptosis is involved in neuron injury, while autophagy is likely to have a protective role.
Collapse
Affiliation(s)
- Lin Qiao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Xindong Xue
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yongyan Shi
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Li Yao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Wanjie Huang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Jun Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Dan Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Na Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Xin Tong
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yanna Du
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yuqing Pan
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
47
|
Nabetani M, Shintaku H, Hamazaki T. Future perspectives of cell therapy for neonatal hypoxic-ischemic encephalopathy. Pediatr Res 2018; 83:356-363. [PMID: 29016557 DOI: 10.1038/pr.2017.260] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 10/05/2017] [Indexed: 12/24/2022]
Abstract
Neonatal ischemic brain injury causes permanent motor-deficit cerebral palsy. Hypoxic-ischemic encephalopathy (HIE) is a very serious condition that can result in death and disability. In 1997, we reported that irreversible neuronal cell damage is induced by the elevation of intracellular Ca ion concentration that has occurred in sequence after excess accumulation of the excitatory neurotransmitter glutamate during ischemia. We also reported that hypothermia was effective in treating ischemic brain damage in rats by suppressing energy loss and raising intracellular Ca ion concentration. Following the 2010 revised International Liaison Committee on Resuscitation guideline, our group developed the Guideline for the treatment of Hypothermia in Japan, and we started online case registry in January 2012. However, therapeutic hypothermia must be initiated within the first 6 h after birth. By contrast, cell therapy may have a much longer therapeutic time window because it might reduce apoptosis/oxidative stress and enhance the regenerative process. In 2014, we administered autologous umbilical cord blood stem cell (UCBC) therapy for neonatal HIE, for the first time in Japan. We enrolled five full-term newborns with moderate-to-severe HIE. Our autologous UCBC therapy is leading to new protocols for the prevention of ischemic brain damage.
Collapse
Affiliation(s)
- Makoto Nabetani
- Department of Pediatrics, Yodogawa Christian Hospital, Osaka, Japan.,Department of Pediatrics, Faculty of Medicine, Osaka City University 1-4-3 Asahi-cho, Abeno-ku, Osaka, Japan
| | - Haruo Shintaku
- Department of Pediatrics, Faculty of Medicine, Osaka City University 1-4-3 Asahi-cho, Abeno-ku, Osaka, Japan
| | - Takashi Hamazaki
- Department of Pediatrics, Faculty of Medicine, Osaka City University 1-4-3 Asahi-cho, Abeno-ku, Osaka, Japan
| |
Collapse
|
48
|
Umbilical cord blood cells for treatment of cerebral palsy; timing and treatment options. Pediatr Res 2018; 83:333-344. [PMID: 28937975 DOI: 10.1038/pr.2017.236] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 09/19/2017] [Indexed: 12/23/2022]
Abstract
Cerebral palsy is the most common cause of physical disability in children, and there is no cure. Umbilical cord blood (UCB) cell therapy for the treatment of children with cerebral palsy is currently being assessed in clinical trials. Although there is much interest in the use of UCB stem cells for neuroprotection and neuroregeneration, the mechanisms of action are not fully understood. Further, UCB contains many stem and progenitor cells of interest, and we will point out that individual cell types within UCB may elicit specific effects. UCB is a clinically proven source of hemotopoietic stem cells (HSCs). It also contains mesenchymal stromal cells (MSCs), endothelial progenitor cells (EPCs), and immunosupressive cells such as regulatory T cells (Tregs) and monocyte-derived supressor cells. Each of these cell types may be individual candidates for the prevention of brain injury following hypoxic and inflammatory events in the perinatal period. We will discuss specific properties of cell types in UCB, with respect to their therapeutic potential and the importance of optimal timing of administration. We propose that tailored cell therapy and targeted timing of administration will optimize the results for future clinical trials in the neuroprotective treatment of perinatal brain injury.
Collapse
|
49
|
From cord to caudate: characterizing umbilical cord blood stem cells and their paracrine interactions with the injured brain. Pediatr Res 2018; 83:205-213. [PMID: 28981488 DOI: 10.1038/pr.2017.251] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 10/02/2017] [Indexed: 01/01/2023]
Abstract
Stem cells are proving to be a promising therapy for a wide range of pediatric disorders, from neonatal hypoxic-ischemic encephalopathy to pediatric leukemia. Owing to their low immunogenicity and ease of availability, umbilical cord blood (UCB) progenitor cells are increasingly replacing fetal- and adult-derived cells in therapeutic settings. Multiple environmental and demographic factors affect the number and type of stem cells extracted from UCB, and these differences have been associated with disparities in outcomes after transplantation. To avoid variations in efficacy, as well as the potential adverse effects of stem cell transplantation, evaluation of the stem cell secretome is critical to identify key paracrine signals released by the stem cells that could be used to provide similar neuroprotective effects to stem cell transplantation. This article describes the cell types found in UCB and reviews the available literature surrounding the effects of collection timing and volume, maternal risk factors, delivery characteristics, and neonatal demographics on the cellular composition of UCB. In addition, the current findings regarding the stem cell secretome are discussed to identify factors that could be used to supplement or replace stem cell transplantation in pediatric neuroprotection.
Collapse
|
50
|
Term vs. preterm cord blood cells for the prevention of preterm brain injury. Pediatr Res 2017; 82:1030-1038. [PMID: 28723885 DOI: 10.1038/pr.2017.170] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/15/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUNDWhite matter brain injury in preterm infants can induce neurodevelopmental deficits. Umbilical cord blood (UCB) cells demonstrate neuroprotective properties, but it is unknown whether cells obtained from preterm cord blood (PCB) vs. term cord blood (TCB) have similar efficacy. This study compared the ability of TCB vs. PCB cells to reduce white matter injury in preterm fetal sheep.METHODSHypoxia-ischemia (HI) was induced in fetal sheep (0.7 gestation) by 25 min umbilical cord occlusion. Allogeneic UCB cells from term or preterm sheep, or saline, were administered to the fetus at 12 h after HI. The fetal brain was collected at 10-day post HI for assessment of white matter neuropathology.RESULTSHI (n=7) induced cell death and microglial activation and reduced total oligodendrocytes and CNPase+myelin protein in the periventricular white matter and internal capsule when compared with control (n=10). Administration of TCB or PCB cells normalized white matter density and reduced cell death and microgliosis (P<0.05). PCB prevented upregulation of plasma tumor necrosis factor (TNF)-a, whereas TCB increased anti-inflammatory interleukin (IL)-10 (P<0.05). TCB, but not PCB, reduced circulating oxidative stress.CONCLUSIONSTCB and PCB cells reduced preterm HI-induced white matter injury, primarily via anti-inflammatory actions. The secondary mechanisms of neuroprotection appear different following TCB vs. PCB administration.
Collapse
|