1
|
Meng H, Huan Y, Zhang K, Yi X, Meng X, Kang E, Wu S, Deng W, Wang Y. Quiescent Adult Neural Stem Cells: Developmental Origin and Regulatory Mechanisms. Neurosci Bull 2024; 40:1353-1363. [PMID: 38656419 PMCID: PMC11365920 DOI: 10.1007/s12264-024-01206-1] [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: 09/26/2023] [Accepted: 01/02/2024] [Indexed: 04/26/2024] Open
Abstract
The existence of neural stem cells (NSCs) in the adult mammalian nervous system, although small in number and restricted to the sub-ventricular zone of the lateral ventricles, the dentate gyrus of the hippocampus, and the olfactory epithelium, is a gift of evolution for the adaptive brain function which requires persistent plastic changes of these regions. It is known that most adult NSCs are latent, showing long cell cycles. In the past decade, the concept of quiescent NSCs (qNSCs) has been widely accepted by researchers in the field, and great progress has been made in the biology of qNSCs. Although the spontaneous neuronal regeneration derived from adult NSCs is not significant, understanding how the behaviors of qNSCs are regulated sheds light on stimulating endogenous NSC-based neuronal regeneration. In this review, we mainly focus on the recent progress of the developmental origin and regulatory mechanisms that maintain qNSCs under normal conditions, and that mobilize qNSCs under pathological conditions, hoping to give some insights for future study.
Collapse
Affiliation(s)
- Han Meng
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yu Huan
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, 110016, China
| | - Kun Zhang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Xuyang Yi
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Xinyu Meng
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
- School of Life Science and Research Center for Natural Peptide Drugs, Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, Yanan University, Yan'an, 716000, China
| | - Enming Kang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Shengxi Wu
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Wenbing Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 510631, China.
| | - Yazhou Wang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Romantsik O, Moreira A, Thébaud B, Ådén U, Ley D, Bruschettini M. Stem cell-based interventions for the prevention and treatment of intraventricular haemorrhage and encephalopathy of prematurity in preterm infants. Cochrane Database Syst Rev 2023; 2:CD013201. [PMID: 36790019 PMCID: PMC9932000 DOI: 10.1002/14651858.cd013201.pub3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
BACKGROUND Germinal matrix-intraventricular haemorrhage (GMH-IVH) and encephalopathy of prematurity (EoP) remain substantial issues in neonatal intensive care units worldwide. Current therapies to prevent or treat these conditions are limited. Stem cell-based therapies offer a potential therapeutic approach to repair, restore, or regenerate injured brain tissue. These preclinical findings have now culminated in ongoing human neonatal studies. This is an update of the 2019 review, which did not include EoP. OBJECTIVES To evaluate the benefits and harms of stem cell-based interventions for prevention or treatment of GM-IVH and EoP in preterm infants. SEARCH METHODS We used standard, extensive Cochrane search methods. The latest search was April 2022. SELECTION CRITERIA We attempted to include randomised controlled trials, quasi-randomised controlled trials, and cluster trials comparing 1. stem cell-based interventions versus control; 2. mesenchymal stromal cells (MSCs) of type or source versus MSCs of other type or source; 3. stem cell-based interventions other than MSCs of type or source versus stem cell-based interventions other than MSCs of other type or source; or 4. MSCs versus stem cell-based interventions other than MSCs. For prevention studies, we included extremely preterm infants (less than 28 weeks' gestation), 24 hours of age or less, without ultrasound diagnosis of GM-IVH or EoP; for treatment studies, we included preterm infants (less than 37 weeks' gestation), of any postnatal age, with ultrasound diagnosis of GM-IVH or with EoP. DATA COLLECTION AND ANALYSIS We used standard Cochrane methods. Our primary outcomes were 1. all-cause neonatal mortality, 2. major neurodevelopmental disability, 3. GM-IVH, 4. EoP, and 5. extension of pre-existing non-severe GM-IVH or EoP. We planned to use GRADE to assess certainty of evidence for each outcome. MAIN RESULTS We identified no studies that met our inclusion criteria. Three studies are currently registered and ongoing. Phase 1 trials are described in the 'Excluded studies' section. AUTHORS' CONCLUSIONS No evidence is currently available to evaluate the benefits and harms of stem cell-based interventions for treatment or prevention of GM-IVH or EoP in preterm infants. We identified three ongoing studies, with a sample size range from 20 to 200. In two studies, autologous cord blood mononuclear cells will be administered to extremely preterm infants via the intravenous route; in one, intracerebroventricular injection of MSCs will be administered to preterm infants up to 34 weeks' gestational age.
Collapse
Affiliation(s)
- Olga Romantsik
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | - Alvaro Moreira
- Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Bernard Thébaud
- Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Canada
- Ottawa Hospital Research Institute, Sprott Centre for Stem Cell Research, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Ulrika Ådén
- Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - David Ley
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | - Matteo Bruschettini
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Lund University, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
5
|
Pedroza-García KA, Calderón-Vallejo D, Quintanar JL. Neonatal Hypoxic-Ischemic Encephalopathy: Perspectives of Neuroprotective and Neuroregenerative Treatments. Neuropediatrics 2022; 53:402-417. [PMID: 36030792 DOI: 10.1055/s-0042-1755235] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a serious condition that could have deleterious neurological outcomes, such as cerebral palsy, neuromotor disability, developmental disability, epilepsy, and sensitive or cognitive problems, and increase the risk of death in severe cases. Once HIE occurs, molecular cascades are triggered favoring the oxidative stress, excitotoxicity, and inflammation damage that promote cell death via apoptosis or necrosis. Currently, the therapeutic hypothermia is the standard of care in HIE; however, it has a small window of action and only can be used in children of more than 36 gestational weeks; for this reason, it is very important to develop new therapies to prevent the progression of the hypoxic-ischemic injury or to develop neuroregenerative therapies in severe HIE cases. The objective of this revision is to describe the emerging treatments for HIE, either preventing cell death for oxidative stress, excitotoxicity, or exacerbated inflammation, as well as describing a new therapeutic approach for neuroregeneration, such as mesenchymal stem cells, brain-derived neurotrophic factor, and gonadotropin realizing hormone agonists.
Collapse
Affiliation(s)
- Karina A Pedroza-García
- Departamento de Fisiología y Farmacología, Laboratorio de Neurofisiología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México
| | - Denisse Calderón-Vallejo
- Departamento de Fisiología y Farmacología, Laboratorio de Neurofisiología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México.,Departamento de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México
| | - J Luis Quintanar
- Departamento de Fisiología y Farmacología, Laboratorio de Neurofisiología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México
| |
Collapse
|
6
|
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
|
7
|
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: 19] [Impact Index Per Article: 6.3] [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
|
8
|
Liu H, Wei T, Huang Q, Liu W, Yang Y, Jin Y, Wu D, Yuan K, Zhang P. The roles, mechanism, and mobilization strategy of endogenous neural stem cells in brain injury. Front Aging Neurosci 2022; 14:924262. [PMID: 36062152 PMCID: PMC9428262 DOI: 10.3389/fnagi.2022.924262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Brain injury poses a heavy disease burden in the world, resulting in chronic deficits. Therapies for brain injuries have been focused on pharmacologic, small molecule, endocrine and cell-based therapies. Endogenous neural stem cells (eNSCs) are a group of stem cells which can be activated in vivo by damage, neurotrophic factors, physical factor stimulation, and physical exercise. The activated eNSCs can proliferate, migrate and differentiate into neuron, oligodendrocyte and astrocyte, and play an important role in brain injury repair and neural plasticity. The roles of eNSCs in the repair of brain injury include but are not limited to ameliorating cognitive function, improving learning and memory function, and promoting functional gait behaviors. The activation and mobilization of eNSCs is important to the repair of injured brain. In this review we describe the current knowledge of the common character of brain injury, the roles and mechanism of eNSCs in brain injury. And then we discuss the current mobilization strategy of eNSCs following brain injury. We hope that a comprehensive awareness of the roles and mobilization strategy of eNSCs in the repair of cerebral ischemia may help to find some new therapeutic targets and strategy for treatment of stroke.
Collapse
Affiliation(s)
- Haijing Liu
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Tao Wei
- Library, Kunming Medical University, Kunming, China
- School of Continuing Education, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Qin Huang
- Department of Teaching Affairs and Administration, Kunming Medical University, Kunming, China
| | - Wei Liu
- School of Public Health, Kunming Medical University, Kunming, China
| | - Yaopeng Yang
- Department of Pulmonary and Critical Care Medicine, The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, China
| | - Yaju Jin
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Danli Wu
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Kai Yuan
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Pengyue Zhang
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| |
Collapse
|
9
|
Effect of expansion of human umbilical cord blood CD34 + cells on neurotrophic and angiogenic factor expression and function. Cell Tissue Res 2022; 388:117-132. [PMID: 35106623 PMCID: PMC8976778 DOI: 10.1007/s00441-022-03592-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 01/19/2022] [Indexed: 12/29/2022]
Abstract
The use of CD34 + cell-based therapies has largely been focused on haematological conditions. However, there is increasing evidence that umbilical cord blood (UCB) CD34 + -derived cells have neuroregenerative properties. Due to low cell numbers of CD34 + cells present in UCB, expansion is required to produce sufficient cells for therapeutic purposes, especially in adults or when frequent applications are required. However, it is not known whether expansion of CD34 + cells has an impact on their function and neuroregenerative capacity. We addressed this knowledge gap in this study, via expansion of UCB-derived CD34 + cells using combinations of LDL, UM171 and SR-1 to yield large numbers of cells and then tested their functionality. CD34 + cells expanded for 14 days in media containing UM171 and SR-1 resulted in over 1000-fold expansion. The expanded cells showed an up-regulation of the neurotrophic factor genes BDNF, GDNF, NTF-3 and NTF-4, as well as the angiogenic factors VEGF and ANG. In vitro functionality testing showed that these expanded cells promoted angiogenesis and, in brain glial cells, promoted cell proliferation and reduced production of reactive oxygen species (ROS) during oxidative stress. Collectively, this study showed that our 14-day expansion protocol provided a robust expansion that could produce enough cells for therapeutic purposes. These expanded cells, when tested in in vitro, maintained functionality as demonstrated through promotion of cell proliferation, attenuation of ROS production caused by oxidative stress and promotion of angiogenesis.
Collapse
|
10
|
Safety of sibling cord blood cell infusion for children with cerebral palsy. Cytotherapy 2022; 24:931-939. [DOI: 10.1016/j.jcyt.2022.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/11/2022] [Accepted: 01/21/2022] [Indexed: 11/23/2022]
|
11
|
Bammidi S, Bali P, Kalra J, Anand A. Transplantation Efficacy of Human Ciliary Epithelium Cells from Fetal Eye and Lin-ve Stem Cells from Umbilical Cord Blood in the Murine Retinal Degeneration Model of Laser Injury. Cell Transplant 2021; 29:963689720946031. [PMID: 33023312 PMCID: PMC7784603 DOI: 10.1177/0963689720946031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A number of degenerative conditions affecting the neural retina including age-related macular degeneration have no successful treatment, resulting in partial or complete vision loss. There are a number of stem cell replacement strategies for recovery of retinal damage using cells from variable sources. However, literature is still deficit in the comparison of efficacy of types of stem cells. The purpose of the study was to compare the therapeutic efficacy of undifferentiated cells, i.e., lineage negative stem cells (Lin-ve SC) with differentiated neurosphere derived from ciliary epithelium (CE) cells on retinal markers associated with laser-induced retinal injury. Laser-induced photocoagulation was carried out to disrupt Bruch’s membrane and retinal pigmented epithelium in C57BL/6 mouse model. Lineage negative cells were isolated from human umbilical cord blood, whereas neurospheres were derived from CE of post-aborted human eyeballs. The cells were then transplanted into subretinal space to study their effect on injury. Markers of neurotropic factors, retina, apoptosis, and proliferation were analyzed after injury and transplantation. mRNA expression was also analyzed by real-time polymerase chain reaction at 1 week, and 3-month immunohistochemistry was evaluated at 1-week time point. CE cell transplantation showed enhanced differentiation of rods and retinal glial cells. However, Lin-ve cells exerted paracrine-dependent modulation of neurotrophic factors, which is possibly mediated by antiapoptotic and proliferative effects. In conclusion, CE transplantation showed superior regenerative outcome in comparison to Lin-ve SC for rescue of artificially injured rodent retinal cells. It is imperative that this source for transplantation may be extensively studied in various doses and additional retinal degeneration models for prospective clinical applications.
Collapse
Affiliation(s)
- Sridhar Bammidi
- Neuroscience Research Lab, Department of Neurology, 29751Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Parul Bali
- Neuroscience Research Lab, Department of Neurology, 29751Post Graduate Institute of Medical Education and Research, Chandigarh, India.,Department of Biophysics, 29751Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Jaswinder Kalra
- Department of Obstetrics and Gynaecology, 29751Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Akshay Anand
- Neuroscience Research Lab, Department of Neurology, 29751Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Yates N, Gunn AJ, Bennet L, Dhillon SK, Davidson JO. Preventing Brain Injury in the Preterm Infant-Current Controversies and Potential Therapies. Int J Mol Sci 2021; 22:1671. [PMID: 33562339 PMCID: PMC7915709 DOI: 10.3390/ijms22041671] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/12/2022] Open
Abstract
Preterm birth is associated with a high risk of morbidity and mortality including brain damage and cerebral palsy. The development of brain injury in the preterm infant may be influenced by many factors including perinatal asphyxia, infection/inflammation, chronic hypoxia and exposure to treatments such as mechanical ventilation and corticosteroids. There are currently very limited treatment options available. In clinical trials, magnesium sulfate has been associated with a small, significant reduction in the risk of cerebral palsy and gross motor dysfunction in early childhood but no effect on the combined outcome of death or disability, and longer-term follow up to date has not shown improved neurological outcomes in school-age children. Recombinant erythropoietin has shown neuroprotective potential in preclinical studies but two large randomized trials, in extremely preterm infants, of treatment started within 24 or 48 h of birth showed no effect on the risk of severe neurodevelopmental impairment or death at 2 years of age. Preclinical studies have highlighted a number of promising neuroprotective treatments, such as therapeutic hypothermia, melatonin, human amnion epithelial cells, umbilical cord blood and vitamin D supplementation, which may be useful at reducing brain damage in preterm infants. Moreover, refinements of clinical care of preterm infants have the potential to influence later neurological outcomes, including the administration of antenatal and postnatal corticosteroids and more accurate identification and targeted treatment of seizures.
Collapse
Affiliation(s)
- Nathanael Yates
- The Queensland Brain Institute, University of Queensland, St Lucia, QLD 4072, Australia;
- School of Human Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | - Alistair J. Gunn
- The Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (A.J.G.); (L.B.); (S.K.D.)
| | - Laura Bennet
- The Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (A.J.G.); (L.B.); (S.K.D.)
| | - Simerdeep K. Dhillon
- The Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (A.J.G.); (L.B.); (S.K.D.)
| | - Joanne O. Davidson
- The Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (A.J.G.); (L.B.); (S.K.D.)
| |
Collapse
|
15
|
Qiu H, Qian T, Wu T, Wang X, Zhu C, Chen C, Wang L. Umbilical cord blood cells for the treatment of preterm white matter injury: Potential effects and treatment options. J Neurosci Res 2020; 99:778-792. [PMID: 33207392 DOI: 10.1002/jnr.24751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/31/2022]
Abstract
Preterm birth is a global public health problem. A large number of preterm infants survive with preterm white matter injury (PWMI), which leads to neurological deficits, and has multifaceted etiology, clinical course, monitoring, and outcomes. The principal upstream insults leading to PWMI initiation are hypoxia-ischemia and infection and/or inflammation and the key target cells are late oligodendrocyte precursor cells. Current PWMI treatments are mainly supportive, and thus have little effect in terms of protecting the immature brain or repairing injury to improve long-term outcomes. Umbilical cord blood (UCB) cells comprise abundant immunomodulatory and stem cells, which have the potential to reduce brain injury, mainly due to anti-inflammatory and immunomodulatory mechanisms, and also through their release of neurotrophic or growth factors to promote endogenous neurogenesis. In this review, we briefly summarize PWMI pathogenesis and pathophysiology, and the specific properties of different cell types in UCB. We further explore the potential mechanism by which UCB can be used to treat PWMI, and discuss the advantages of and potential issues related to UCB cell therapy. Finally, we suggest potential future studies of UCB cell therapy in preterm infants.
Collapse
Affiliation(s)
- Han Qiu
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| | - Tianyang Qian
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| | - Tong Wu
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| | - Xiaoyang Wang
- Center of Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Changlian Zhu
- Center of Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Chao Chen
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| | - Laishuan Wang
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
16
|
Markosyan V, Safiullov Z, Izmailov A, Fadeev F, Sokolov M, Kuznetsov M, Trofimov D, Kim E, Kundakchyan G, Gibadullin A, Salafutdinov I, Nurullin L, Bashirov F, Islamov R. Preventive Triple Gene Therapy Reduces the Negative Consequences of Ischemia-Induced Brain Injury after Modelling Stroke in a Rat. Int J Mol Sci 2020; 21:ijms21186858. [PMID: 32962079 PMCID: PMC7558841 DOI: 10.3390/ijms21186858] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
Currently, the main fundamental and clinical interest for stroke therapy is focused on developing a neuroprotective treatment of a penumbra region within the therapeutic window. The development of treatments for ischemic stroke in at-risk patients is of particular interest. Preventive gene therapy may significantly reduce the negative consequences of ischemia-induced brain injury. In the present study, we suggest the approach of preventive gene therapy for stroke. Adenoviral vectors carrying genes encoding vascular endothelial growth factor (VEGF), glial cell-derived neurotrophic factor (GDNF) and neural cell adhesion molecule (NCAM) or gene engineered umbilical cord blood mononuclear cells (UCB-MC) overexpressing recombinant VEGF, GDNF, and NCAM were intrathecally injected before distal occlusion of the middle cerebral artery in rats. Post-ischemic brain recovery was investigated 21 days after stroke modelling. Morphometric and immunofluorescent analysis revealed a reduction of infarction volume accompanied with a lower number of apoptotic cells and decreased expression of Hsp70 in the peri-infarct region in gene-treated animals. The lower immunopositive areas for astrocytes and microglial cells markers, higher number of oligodendrocytes and increased expression of synaptic proteins suggest the inhibition of astrogliosis, supporting the corresponding myelination and functional recovery of neurons in animals receiving preventive gene therapy. In this study, for the first time, we provide evidence of the beneficial effects of preventive triple gene therapy by an adenoviral- or UCB-MC-mediated intrathecal simultaneous delivery combination of vegf165, gdnf, and ncam1 on the preservation and recovery of the brain in rats with subsequent modelling of stroke.
Collapse
Affiliation(s)
- Vage Markosyan
- Department of Medical Biology and Genetics, Kazan State Medical University, 420012 Kazan, Russia; (V.M.); (Z.S.); (A.I.); (F.F.); (M.S.); (M.K.); (D.T.); (E.K.); (A.G.); (F.B.)
| | - Zufar Safiullov
- Department of Medical Biology and Genetics, Kazan State Medical University, 420012 Kazan, Russia; (V.M.); (Z.S.); (A.I.); (F.F.); (M.S.); (M.K.); (D.T.); (E.K.); (A.G.); (F.B.)
| | - Andrei Izmailov
- Department of Medical Biology and Genetics, Kazan State Medical University, 420012 Kazan, Russia; (V.M.); (Z.S.); (A.I.); (F.F.); (M.S.); (M.K.); (D.T.); (E.K.); (A.G.); (F.B.)
| | - Filip Fadeev
- Department of Medical Biology and Genetics, Kazan State Medical University, 420012 Kazan, Russia; (V.M.); (Z.S.); (A.I.); (F.F.); (M.S.); (M.K.); (D.T.); (E.K.); (A.G.); (F.B.)
| | - Mikhail Sokolov
- Department of Medical Biology and Genetics, Kazan State Medical University, 420012 Kazan, Russia; (V.M.); (Z.S.); (A.I.); (F.F.); (M.S.); (M.K.); (D.T.); (E.K.); (A.G.); (F.B.)
| | - Maksim Kuznetsov
- Department of Medical Biology and Genetics, Kazan State Medical University, 420012 Kazan, Russia; (V.M.); (Z.S.); (A.I.); (F.F.); (M.S.); (M.K.); (D.T.); (E.K.); (A.G.); (F.B.)
| | - Dmitry Trofimov
- Department of Medical Biology and Genetics, Kazan State Medical University, 420012 Kazan, Russia; (V.M.); (Z.S.); (A.I.); (F.F.); (M.S.); (M.K.); (D.T.); (E.K.); (A.G.); (F.B.)
| | - Evgeny Kim
- Department of Medical Biology and Genetics, Kazan State Medical University, 420012 Kazan, Russia; (V.M.); (Z.S.); (A.I.); (F.F.); (M.S.); (M.K.); (D.T.); (E.K.); (A.G.); (F.B.)
| | - Grayr Kundakchyan
- Institute of Fundamental Medicine and Biology, Kazan [Volga Region] Federal University, 420008 Kazan, Russia; (G.K.); (I.S.)
| | - Airat Gibadullin
- Department of Medical Biology and Genetics, Kazan State Medical University, 420012 Kazan, Russia; (V.M.); (Z.S.); (A.I.); (F.F.); (M.S.); (M.K.); (D.T.); (E.K.); (A.G.); (F.B.)
| | - Ilnur Salafutdinov
- Institute of Fundamental Medicine and Biology, Kazan [Volga Region] Federal University, 420008 Kazan, Russia; (G.K.); (I.S.)
| | - Leniz Nurullin
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center of Kazan Scientific Center of Russian Academy of Sciences, 119991 Kazan, Russia;
| | - Farid Bashirov
- Department of Medical Biology and Genetics, Kazan State Medical University, 420012 Kazan, Russia; (V.M.); (Z.S.); (A.I.); (F.F.); (M.S.); (M.K.); (D.T.); (E.K.); (A.G.); (F.B.)
| | - Rustem Islamov
- Department of Medical Biology and Genetics, Kazan State Medical University, 420012 Kazan, Russia; (V.M.); (Z.S.); (A.I.); (F.F.); (M.S.); (M.K.); (D.T.); (E.K.); (A.G.); (F.B.)
- Correspondence:
| |
Collapse
|
17
|
Bruschettini M, Romantsik O, Moreira A, Ley D, Thébaud B. Stem cell-based interventions for the prevention of morbidity and mortality following hypoxic-ischaemic encephalopathy in newborn infants. Cochrane Database Syst Rev 2020; 8:CD013202. [PMID: 32813884 PMCID: PMC7438027 DOI: 10.1002/14651858.cd013202.pub2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hypoxic-ischaemic encephalopathy (HIE) is a leading cause of mortality and long-term neurological sequelae, affecting thousands of children worldwide. Current therapies to treat HIE are limited to cooling. Stem cell-based therapies offer a potential therapeutic approach to repair or regenerate injured brain tissue. These preclinical findings have now culminated in ongoing human neonatal trials. OBJECTIVES To determine the efficacy and safety of stem cell-based interventions for the treatment of hypoxic-ischaemic encephalopathy (HIE) in newborn infants. SEARCH METHODS We used the standard search strategy of Cochrane Neonatal to search the Cochrane Central Register of Controlled Trials (CENTRAL; 2020, Issue 5), MEDLINE via PubMed (1966 to 8 June 2020), Embase (1980 to 8 June 2020), and CINAHL (1982 to 8 June 2020). We also searched clinical trials databases, conference proceedings, and the reference lists of retrieved articles for randomised controlled trials and quasi-randomised trials. SELECTION CRITERIA Randomised controlled trials, quasi-randomised controlled trials and cluster trials comparing 1) stem cell-based interventions (any type) compared to control (placebo or no treatment); 2) use of mesenchymal stem/stromal cells (MSCs) of type (e.g. number of doses or passages) or source (e.g. autologous versus allogeneic, or bone marrow versus cord) versus MSCs of other type or source; 3) use of stem cell-based interventions other than MSCs of type (e.g. mononuclear cells, oligodendrocyte progenitor cells, neural stem cells, hematopoietic stem cells, and inducible pluripotent stem cells) or source (e.g. autologous versus allogeneic, or bone marrow versus cord) versus stem cell-based interventions other than MSCs of other type or source; or 4) MSCs versus stem cell-based interventions other than MSCs. DATA COLLECTION AND ANALYSIS For each of the included trials, two authors independently planned to extract data (e.g. number of participants, birth weight, gestational age, type and source of MSCs or other stem cell-based interventions) and assess the risk of bias (e.g. adequacy of randomisation, blinding, completeness of follow-up). The primary outcomes considered in this review are all-cause neonatal mortality, major neurodevelopmental disability, death or major neurodevelopmental disability assessed at 18 to 24 months of age. We planned to use the GRADE approach to assess the quality of evidence. MAIN RESULTS Our search strategy yielded 616 references. Two review authors independently assessed all references for inclusion. We did not find any completed studies for inclusion. Fifteen RCTs are currently registered and ongoing. We describe the three studies we excluded. AUTHORS' CONCLUSIONS There is currently no evidence from randomised trials that assesses the benefit or harms of stem cell-based interventions for the prevention of morbidity and mortality following hypoxic-ischaemic encephalopathy in newborn infants.
Collapse
Affiliation(s)
- Matteo Bruschettini
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Lund University, Skåne University Hospital, Lund, Sweden
| | - Olga Romantsik
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | - Alvaro Moreira
- Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - David Ley
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | - Bernard Thébaud
- Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Canada
- Ottawa Hospital Research Institute, Sprott Centre for Stem Cell Research, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| |
Collapse
|
18
|
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
|
19
|
Romantsik O, Bruschettini M, Moreira A, Thébaud B, Ley D. Stem cell-based interventions for the prevention and treatment of germinal matrix-intraventricular haemorrhage in preterm infants. Cochrane Database Syst Rev 2019; 9:CD013201. [PMID: 31549743 PMCID: PMC6757514 DOI: 10.1002/14651858.cd013201.pub2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Germinal matrix-intraventricular haemorrhage (GMH-IVH) remains a substantial issue in neonatal intensive care units worldwide. Current therapies to prevent or treat GMH-IVH are limited. Stem cell-based therapies offer a potential therapeutic approach to repair, restore, and/or regenerate injured brain tissue. These preclinical findings have now culminated in ongoing human neonatal studies. OBJECTIVES To determine the benefits and harms of stem cell-based interventions for prevention or treatment of germinal matrix-intraventricular haemorrhage (GM-IVH) in preterm infants. SEARCH METHODS We used the standard search strategy of Cochrane Neonatal to search the Cochrane Central Register of Controlled Trials (CENTRAL; 2019, Issue 1), in the Cochrane Library; MEDLINE via PubMed (1966 to 7 January 2019); Embase (1980 to 7 January 2019); and the Cumulative Index to Nursing and Allied Health Literature (CINAHL) (1982 to 7 January 2019). We also searched clinical trials databases, conference proceedings, and reference lists of retrieved articles for randomised controlled trials and quasi-randomised trials. SELECTION CRITERIA We attempted to identify randomised controlled trials, quasi-randomised controlled trials, and cluster trials comparing (1) stem cell-based interventions versus control; (2) mesenchymal stromal cells (MSCs) of type or source versus MSCs of other type or source; (3) stem cell-based interventions other than MSCs of type or source versus stem cell-based interventions other than MSCs of other type or source; or (4) MSCs versus stem cell-based interventions other than MSCs. For prevention studies, we included extremely preterm infants (less than 28 weeks' gestation), 24 hours of age or less, without ultrasound diagnosis of GM-IVH; for treatment studies, we included preterm infants (less than 37 weeks' gestation), of any postnatal age, with ultrasound diagnosis of GM-IVH. DATA COLLECTION AND ANALYSIS For each of the included trials, two review authors independently planned to extract data (e.g. number of participants, birth weight, gestational age, type and source of MSCs, other stem cell-based interventions) and assess the risk of bias (e.g. adequacy of randomisation, blinding, completeness of follow-up). Primary outcomes considered in this review are all-cause neonatal mortality, major neurodevelopmental disability, GM-IVH, and extension of pre-existing non-severe GM-IVH. We planned to use the GRADE approach to assess the quality of evidence. MAIN RESULTS Our search strategy yielded 769 references. We did not find any completed studies for inclusion. One randomised controlled trial is currently registered and ongoing. Five phase 1 trials are described in the excluded studies. AUTHORS' CONCLUSIONS Currently no evidence is available to show the benefits or harms of stem cell-based interventions for treatment or prevention of GM-IVH in preterm infants.
Collapse
Affiliation(s)
- Olga Romantsik
- Lund University, Skåne University HospitalDepartment of Clinical Sciences Lund, PaediatricsLundSweden
| | - Matteo Bruschettini
- Lund University, Skåne University HospitalDepartment of Clinical Sciences Lund, PaediatricsLundSweden
- Skåne University HospitalCochrane SwedenWigerthuset, Remissgatan 4, first floorroom 11‐221LundSweden22185
| | - Alvaro Moreira
- University of Texas Health Science Center at San AntonioPediatrics, Division of NeonatologySan AntonioTexasUSA
| | - Bernard Thébaud
- Children's Hospital of Eastern OntarioDepartment of PediatricsOttawaONCanada
- Ottawa Hospital Research Institute, Sprott Centre for Stem Cell ResearchOttawaCanada
- University of OttawaDepartment of Cellular and Molecular MedicineOttawaCanada
| | - David Ley
- Lund University, Skane University HospitalDepartment of Clinical Sciences Lund, PaediatricsLundSweden
| | | |
Collapse
|
20
|
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
|
21
|
Lin X, Li J, Xing YQ. Geniposide, a sonic hedgehog signaling inhibitor, inhibits the activation of hepatic stellate cell. Int Immunopharmacol 2019; 72:330-338. [DOI: 10.1016/j.intimp.2019.04.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 04/06/2019] [Accepted: 04/07/2019] [Indexed: 11/27/2022]
|
22
|
Zhang J, Yang C, Chen J, Luo M, Qu Y, Mu D, Chen Q. Umbilical cord mesenchymal stem cells and umbilical cord blood mononuclear cells improve neonatal rat memory after hypoxia-ischemia. Behav Brain Res 2019; 362:56-63. [PMID: 30639506 DOI: 10.1016/j.bbr.2019.01.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/06/2019] [Accepted: 01/08/2019] [Indexed: 12/26/2022]
Abstract
Current treatment options for hypoxic-ischemic encephalopathy (HIE) are limited. Human umbilical cord mesenchymal stem cells (UC-MSCs) and cord blood mononuclear cells (CB-MNCs) offer great potential for the treatment of many neurological diseases. The aim of the present study was to identify which cell type is more effective for the treatment of HIE. PKH26-labeled UC-MSCs and CB-MNCs were transplanted into rats with hypoxia-ischemia (HI)-induced brain damage. Apoptotic cell numbers in the brain, as labeled by TUNEL, were assessed. Myelination and gliosis were investigated using myelin basic protein and glial fibrillary acidic protein immunohistochemistry, respectively. The Morris water maze was used to assess animal learning abilities. Our data show that transplantation of UC-MSCs or CB-MNCs after HI reduced astrogliosis, prevented neuronal loss in the striatum, and markedly improved functional brain outcomes after a 28-day recovery period. Moreover, treatment with CB-MNCs increased the proportion of mature oligodendrocytes and improved myelination in cortical areas. Both UC-MSCs and CB-MNCs may result in the recovery of neurological function in HI rats. Based on our data, UC-MSCs and UCB-MNCs may be particularly effective stem cell subsets for treatment of neonatal HIE.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chao Yang
- Stem Cells and Regenerative Medicine Research Center, Sichuan Cord Blood Bank/Sichuan Neo-Life Stem Cell Biotech Inc., Chengdu, 610037, Sichuan, China
| | - Juan Chen
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Maowen Luo
- Stem Cells and Regenerative Medicine Research Center, Sichuan Cord Blood Bank/Sichuan Neo-Life Stem Cell Biotech Inc., Chengdu, 610037, Sichuan, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu, 610041, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu, 610041, China
| | - Qiang Chen
- Stem Cells and Regenerative Medicine Research Center, Sichuan Cord Blood Bank/Sichuan Neo-Life Stem Cell Biotech Inc., Chengdu, 610037, Sichuan, China
| |
Collapse
|
23
|
Spiliopoulos S, Festas G, Reppas L, Brountzos E. Intra-arterial administration of cell-based biological agents for ischemic stroke therapy. Expert Opin Biol Ther 2019; 19:249-259. [PMID: 30615496 DOI: 10.1080/14712598.2019.1566454] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/05/2019] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Ischemic stroke is becoming a primary cause of disability and death worldwide. To date, therapeutic options remain limited focusing on mechanical thrombolysis or administration of thrombolytic agents. However, these therapies do not promote neuroprotection and neuro-restoration of the ischemic area of the brain. AREAS COVERED This review highlights the option of minimal invasive, intra-arterial, administration of biological agents for stroke therapy. The authors provide an update of all available studies, discuss issues that influence outcomes and describe future perspectives which aim to improve clinical outcomes. New therapeutic options based on cellular and molecular interactions following an ischemic brain event, will be highlighted. EXPERT OPINION Intra-arterial administration of biological agents during trans-catheter thrombolysis or thrombectomy could limit neuronal cell death and facilitate regeneration or neurogenesis following ischemic brain injury. Despite the initial progress, further meticulous studies are needed in order to establish the clinical use of stem cell-induced neuroprotection and neuroregeneration.
Collapse
Affiliation(s)
- Stavros Spiliopoulos
- a 2nd Department of Radiology, Division of Interventional Radiology, School of Medicine , National and Kapodistrian University of Athens, Attikon University Hospital , Athens , Greece
| | - Georgios Festas
- a 2nd Department of Radiology, Division of Interventional Radiology, School of Medicine , National and Kapodistrian University of Athens, Attikon University Hospital , Athens , Greece
| | - Lazaros Reppas
- a 2nd Department of Radiology, Division of Interventional Radiology, School of Medicine , National and Kapodistrian University of Athens, Attikon University Hospital , Athens , Greece
| | - Elias Brountzos
- a 2nd Department of Radiology, Division of Interventional Radiology, School of Medicine , National and Kapodistrian University of Athens, Attikon University Hospital , Athens , Greece
| |
Collapse
|
24
|
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
|
25
|
Bruschettini M, Romantsik O, Moreira A, Ley D, Thébaud B. Stem cell-based interventions for the prevention of morbidity and mortality following hypoxic-ischaemic encephalopathy in newborn infants. Hippokratia 2018. [DOI: 10.1002/14651858.cd013202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Matteo Bruschettini
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
- Skåne University Hospital; Cochrane Sweden; Wigerthuset, Remissgatan 4, first floor room 11-221 Lund Sweden 22185
| | - Olga Romantsik
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
| | - Alvaro Moreira
- University of Texas Health Science Center at San Antonio; Pediatrics, Division of Neonatology; San Antonio Texas USA
| | - David Ley
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
| | - Bernard Thébaud
- Children's Hospital of Eastern Ontario; Department of Pediatrics; Ottawa ON Canada
- Ottawa Hospital Research Institute, Sprott Center for Stem Cell Research; Ottawa Canada
- University of Ottawa; Department of Cellular and Molecular Medicine; Ottawa Canada
| |
Collapse
|
26
|
Romantsik O, Bruschettini M, Moreira A, Thébaud B, Ley D. Stem cell-based interventions for the prevention and treatment of germinal matrix-intraventricular haemorrhage in preterm infants. Hippokratia 2018. [DOI: 10.1002/14651858.cd013201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Olga Romantsik
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
| | - Matteo Bruschettini
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
- Skåne University Hospital; Cochrane Sweden; Wigerthuset, Remissgatan 4, first floor room 11-221 Lund Sweden 22185
| | - Alvaro Moreira
- University of Texas Health Science Center at San Antonio; Pediatrics, Division of Neonatology; San Antonio Texas USA
| | - Bernard Thébaud
- Children's Hospital of Eastern Ontario; Department of Pediatrics; Ottawa ON Canada
- Ottawa Hospital Research Institute, Sprott Center for Stem Cell Research; Ottawa Canada
- University of Ottawa; Department of Cellular and Molecular Medicine; Ottawa Canada
| | - David Ley
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
| |
Collapse
|
27
|
The Neuroprotective Roles of Sonic Hedgehog Signaling Pathway in Ischemic Stroke. Neurochem Res 2018; 43:2199-2211. [DOI: 10.1007/s11064-018-2645-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 07/29/2018] [Accepted: 09/19/2018] [Indexed: 01/20/2023]
|
28
|
Sokolov ME, Bashirov FV, Markosyan VA, Povysheva TV, Fadeev FO, Izmailov AA, Kuztetsov MS, Safiullov ZZ, Shmarov MM, Naroditskyi BS, Palotás A, Islamov RR. Triple-Gene Therapy for Stroke: A Proof-of-Concept in Vivo Study in Rats. Front Pharmacol 2018; 9:111. [PMID: 29497380 PMCID: PMC5818439 DOI: 10.3389/fphar.2018.00111] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 01/31/2018] [Indexed: 12/26/2022] Open
Abstract
Natural brain repair after stroke is extremely limited, and current therapeutic options are even more scarce with no clinical break-through in sight. Despite restricted regeneration in the central nervous system, we have previously proved that human umbilical cord blood mono-nuclear cells (UCB-MC) transduced with adenoviral vectors carrying genes encoding vascular endothelial growth factor (VEGF), glial cell-derived neurotrophic factor (GDNF), and neural cell adhesion molecule (NCAM) successfully rescued neurons in amyotrophic lateral sclerosis and spinal cord injury. This proof-of-principle project was aimed at evaluating the beneficial effects of the same triple-gene approach in stroke. Rats subjected to distal occlusion of the middle cerebral artery were treated intrathecally with a combination of these genes either directly or using our cell-based (UCB-MC) approach. Various techniques and markers were employed to evaluate brain injury and subsequent recovery after treatment. Brain repair was most prominent when therapeutic genes were delivered via adenoviral vector- or UCB-MC-mediated approach. Remodeling of brain cortex in the stroke area was confirmed by reduction of infarct volume and attenuated neural cell death, depletion of astrocytes and microglial cells, and increase in the number of oligodendroglial cells and synaptic proteins expression. These results imply that intrathecal injection of genetically engineered UCB-MC over-expressing therapeutic molecules (VEGF, GDNF, and NCAM) following cerebral blood vessel occlusion might represent a novel avenue for future research into treating stroke.
Collapse
Affiliation(s)
- Mikhail E Sokolov
- Department of Medical Biology and Genetics, Kazan State Medical University, Kazan, Russia
| | - Farid V Bashirov
- Department of Medical Biology and Genetics, Kazan State Medical University, Kazan, Russia
| | - Vage A Markosyan
- Department of Medical Biology and Genetics, Kazan State Medical University, Kazan, Russia
| | - Tatyana V Povysheva
- Department of Medical Biology and Genetics, Kazan State Medical University, Kazan, Russia
| | - Filip O Fadeev
- Department of Medical Biology and Genetics, Kazan State Medical University, Kazan, Russia
| | - Andrey A Izmailov
- Department of Medical Biology and Genetics, Kazan State Medical University, Kazan, Russia
| | - Maxim S Kuztetsov
- Department of Medical Biology and Genetics, Kazan State Medical University, Kazan, Russia
| | - Zufar Z Safiullov
- Department of Medical Biology and Genetics, Kazan State Medical University, Kazan, Russia
| | - Maxim M Shmarov
- Gamaleya Research Institute of Epidemiology and Microbiology, Moscow, Russia
| | - Boris S Naroditskyi
- Gamaleya Research Institute of Epidemiology and Microbiology, Moscow, Russia
| | - András Palotás
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia.,Asklepios-Med (Private Medical Practice and Research Center), Szeged, Hungary
| | - Rustem R Islamov
- Department of Medical Biology and Genetics, Kazan State Medical University, Kazan, Russia.,Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia.,Kazan Institute of Biochemistry and Biophysics, Russian Academy of Sciences, Kazan, Russia
| |
Collapse
|
29
|
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
|
30
|
Yu X, Wu H, Zhao Y, Guo Y, Chen Y, Dong P, Mu Q, Wang X, Wang X. Bone marrow mesenchymal stromal cells alleviate brain white matter injury via the enhanced proliferation of oligodendrocyte progenitor cells in focal cerebral ischemic rats. Brain Res 2017; 1680:127-136. [PMID: 29258846 DOI: 10.1016/j.brainres.2017.12.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 12/11/2017] [Accepted: 12/13/2017] [Indexed: 02/05/2023]
Abstract
The effects of transplanting bone marrow mesenchymal stromal cells (BMSCs) for the treatment of white matter damage are not well understood, nor are the underlying mechanisms. Recent studies showed that endogenous oligodendrocyte progenitor cells (OPCs) can be stimulated to proliferate. Therefore, we explore the effects of BMSCs transplantation on white matter damage and the proliferation of OPCs in transient focal cerebral ischemic rats. BMSCs were transplanted into a group of rats that had undergone middle cerebral artery occlusion (MCAO) 24 h after reperfusion. The ratswere examined by MRI-T2 and DTI sequencesdynamically. The proliferating cells were labeled by 5-Bromo-2'-deoxyuridine (BrdU). The effects of BMSC transplantation on neurons, axons, myelination, and proliferating OPCs were examined by Nissl staining, MBP/NF-H and BrdU/NG2 immunofluorescence staining7 days after transplantation. More Nissl-stained neuronswere found and the FA value of MRI-DTI was significantly higher in the MCAO + BMSCs group than in the MCAOgroup (both P < .01). The fold change of MBP protein was significantly higher in the MCAO + BMSCs group than in the MCAO group (P < .01); the same was true of NF-H protein. Additionally, there were more BrdU+NG2+ cells in the SVZ areas of the MCAO + BMSCs group than in the MCAO group (P < .01). BMSCs thus were shown to alleviate neuronal/axonal injury and promote the proliferation of OPCs and formation of myelin sheath, significantly alleviating white matter damage in focal cerebral ischemic rats.
Collapse
Affiliation(s)
- Xiaohe Yu
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Hongjuan Wu
- Clinical Medical Institute, Weifang Medical University, Weifang 261053, PR China
| | - Yansong Zhao
- Department of Ophthalmology, Weifang Medical University Affiliated Hospital, Clinical Medical Institute, Weifang Medical University, Weifang 261053, PR China
| | - Yuanyuan Guo
- Department of Medical Imaging, Weifang Medical University, Weifang 261053, PR China
| | - Yuxi Chen
- Department of Medical Imaging, Weifang Medical University, Weifang 261053, PR China
| | - Peng Dong
- Department of Medical Imaging, Weifang Medical University, Weifang 261053, PR China
| | - Qingjie Mu
- Department of Hematology, Clinical Medical Institute, Weifang Medical University, Weifang 261053, PR China
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Xiaoli Wang
- Department of Medical Imaging, Weifang Medical University, Weifang 261053, PR China.
| |
Collapse
|
31
|
Qi C, Zhang J, Chen X, Wan J, Wang J, Zhang P, Liu Y. Hypoxia stimulates neural stem cell proliferation by increasing HIF‑1α expression and activating Wnt/β-catenin signaling. ACTA ACUST UNITED AC 2017; 63:12-19. [PMID: 28838333 DOI: 10.14715/cmb/2017.63.7.2] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Indexed: 01/26/2023]
Abstract
Evidence indicates that after brain injury, neurogenesis is enhanced in regions such as hippocampus, striatum, and cortex. To study the role of hypoxia-inducible factor-1 (HIF‑1α) and Wnt signaling in cerebral ischemia/hypoxia-induced proliferation of neural stem cells (NSCs), we investigated the proliferation of NSCs, expression of HIF‑1α, and activation of Wnt signaling under conditions of pathologic hypoxia in vitro. NSCs were isolated from 30-day-old Sprague-Dawley rats and subjected to 0.3% oxygen in a microaerophilic incubation system. Cell proliferation was evaluated by measuring the diameter of neurospheres and by bromodeoxyuridine incorporation assays. Real-time quantitative PCR and Western blotting were used to detect mRNA and protein levels of HIF-1α, β-catenin, and cyclin D1 in the NSCs. The results showed that hypoxia increased NSC proliferation and the levels of HIF-1α, β‑catenin, and cyclin D1 (p < 0.05). Blockade of the Wnt signaling pathway decreased hypoxia-induced NSC proliferation, whereas activation of this pathway increased hypoxia-induced NSC proliferation (p < 0.05). Knockdown of HIF-1α with HIF-1α siRNA decreased β‑catenin nuclear translocation and cyclin D1 expression, and inhibited proliferation of NSCs (p < 0.05). These findings indicate that pathologic hypoxia stimulates NSC proliferation by increasing expression of HIF-1α and activating the Wnt/β-catenin signaling pathway. The data suggest that Wnt/β-catenin signaling may play a key role in NSC proliferation under conditions of pathologic hypoxia.
Collapse
Affiliation(s)
- C Qi
- Institute of Neurobiology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061 China
| | - J Zhang
- Institute of Neurobiology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061 China
| | - X Chen
- Institute of Neurobiology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061 China
| | - J Wan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, USA
| | - J Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, USA
| | - P Zhang
- Institute of Neurobiology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061 China
| | - Y Liu
- Institute of Neurobiology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061 China
| |
Collapse
|
32
|
Dai Y, Li W, Zhong M, Chen J, Cheng Q, Liu Y, Li T. The paracrine effect of cobalt chloride on BMSCs during cognitive function rescue in the HIBD rat. Behav Brain Res 2017; 332:99-109. [PMID: 28576310 DOI: 10.1016/j.bbr.2017.05.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 05/20/2017] [Accepted: 05/24/2017] [Indexed: 12/22/2022]
Abstract
Hypoxia-ischemia (HI)-induced perinatal encephalopathy frequently causes chronic neurological morbidities and acute mortality. Bone mesenchymal stem cell (BMSC) transplantation could potentially promote functional and anatomical recovery of ischemic tissue. In vitro hypoxic preconditioning is an effective strategy to improve the survival of BMSCs in ischemic tissue. In this study, cobalt chloride (CoCl2) preconditioned medium from BMSC cultures was injected into the left lateral ventricle of HI rats using a micro-osmotic pump at a flow rate 1.0μl/h for 7 days. The protein levels of HIF-1α and its target genes, vascular endothelial growth factor and erythropoietin, markedly increased after CoCl2 preconditioning in BMSCs. In 7-week-old rats that received CoCl2 preconditioned BMSC medium, results of the Morris water maze test indicated ameliorated spatial working memory function following hypoxia-ischemia damage. Neuronal loss, cellular disorganization, and shrinkage in brain tissue were also ameliorated. Extracellular field excitatory postsynaptic potentials (fEPSPs) in the brain slices of 8-week-old rats were recorded; administration of CoCl2 preconditioned BMSC culture medium induced a progressive increment of baseline and amplitude of the fEPSPs. Immunohistochemical quantification showed that GluR2 protein expression increased. In conclusion, CoCl2 activates HIF-1α signals in BMSCs. CoCl2 preconditioned BMSC culture medium likely effects neuroprotection by inducing long-term potentiation (LTP), which could be associated with GluR2 expression. The paracrine effects of hypoxia preconditioning on BMSCs could have applications in novel cell-based therapeutic strategies for hypoxic and ischemic brain injury.
Collapse
MESH Headings
- Animals
- Brain/pathology
- Brain/physiopathology
- Cells, Cultured
- Cobalt/pharmacology
- Culture Media, Conditioned
- Disease Models, Animal
- Excitatory Postsynaptic Potentials
- Female
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Ischemia, Brain/pathology
- Hypoxia-Ischemia, Brain/physiopathology
- Hypoxia-Ischemia, Brain/psychology
- Hypoxia-Ischemia, Brain/therapy
- Male
- Maze Learning/physiology
- Memory, Short-Term/physiology
- Mesenchymal Stem Cell Transplantation
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/physiology
- Neuroprotection/physiology
- Protective Agents/pharmacology
- Rats, Sprague-Dawley
- Receptors, AMPA/metabolism
- Spatial Memory/physiology
- Tissue Culture Techniques
Collapse
Affiliation(s)
- Ying Dai
- Department of Primary Child Health Care, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Wendi Li
- Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China
| | - Min Zhong
- Department of Neurology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Jie Chen
- Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China
| | - Qian Cheng
- Department of Primary Child Health Care, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Youxue Liu
- Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China.
| | - Tingyu Li
- Department of Primary Child Health Care, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China; Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China.
| |
Collapse
|
33
|
Patel SS, Tomar S, Sharma D, Mahindroo N, Udayabanu M. Targeting sonic hedgehog signaling in neurological disorders. Neurosci Biobehav Rev 2017; 74:76-97. [PMID: 28088536 DOI: 10.1016/j.neubiorev.2017.01.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/29/2016] [Accepted: 01/07/2017] [Indexed: 12/13/2022]
Abstract
Sonic hedgehog (Shh) signaling influences neurogenesis and neural patterning during the development of central nervous system. Dysregulation of Shh signaling in brain leads to neurological disorders like autism spectrum disorder, depression, dementia, stroke, Parkinson's diseases, Huntington's disease, locomotor deficit, epilepsy, demyelinating disease, neuropathies as well as brain tumors. The synthesis, processing and transport of Shh ligand as well as the localization of its receptors and signal transduction in the central nervous system has been carefully reviewed. Further, we summarize the regulation of small molecule modulators of Shh pathway with potential in neurological disorders. In conclusion, further studies are warranted to demonstrate the potential of positive and negative regulators of the Shh pathway in neurological disorders.
Collapse
Affiliation(s)
- Sita Sharan Patel
- Department of Pharmacy, Jaypee University of Information Technology, Waknaghat 173234, Himachal Pradesh, India
| | - Sunil Tomar
- School of Pharmaceutical Sciences, Shoolini University, Post Box 9, Solan 173212, Himachal Pradesh, India
| | - Diksha Sharma
- School of Pharmaceutical Sciences, Shoolini University, Post Box 9, Solan 173212, Himachal Pradesh, India
| | - Neeraj Mahindroo
- School of Pharmaceutical Sciences, Shoolini University, Post Box 9, Solan 173212, Himachal Pradesh, India
| | - Malairaman Udayabanu
- Department of Pharmacy, Jaypee University of Information Technology, Waknaghat 173234, Himachal Pradesh, India.
| |
Collapse
|
34
|
[Effects of umbilical cord blood mononuclear cells transplantation via lateral ventricle on the neural apoptosis and the expression of Bax and Bcl-2 proteins in neonatal rats with hypoxic-ischemic brain damage]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2016. [PMID: 27655545 PMCID: PMC7389963 DOI: 10.7499/j.issn.1008-8830.2016.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To explore the effects of umbilical cord blood mononuclear cells (UCBMC) transplantation on the neuronal apoptosis and the expression of Bcl-2 and Bax proteins in neonatal rats with hypoxic-ischemic brain damage (HIBD). METHODS Seven-day-old Sprague-Dawley neonatal rats were randomly divided into normal control (N)+normal saline (NS), HIBD+NS, N+UCBMC, and HIBD+UCBMC groups. HIBD model was prepared using the classical Rice-Vannucci method. Twenty-four hours after HIBD, UCBMC were transplanted in the N+UCBMC and HIBD+UCBMC groups. Seven days after transplantation, NeuN/Cleaved-Caspase-3 double immunofluorescence staining and TUNEL methods were used to observe neural apoptosis in the cortex. The expression levels of Bax and Bcl-2 proteins were examined by Western blot analysis. RESULTS There were more NeuN+ cleaved Caspase-3+DAPI+ and TUNEL+DAPI+ cells in the HIBD+NS group compared with the N+NS and N+UCBMC groups (P<0.01). There were less NeuN+ cleaved Caspase-3+DAPI+ and TUNEL+DAPI+ cells in the HIBD+UCBMC group compared with the HIBD+NS group (P<0.01). The concentration of Bax protein was higher and that of Bcl-2 proteins was lower in the HIBD+NS group compared with the N+NS and N+UCBMC groups (P<0.01). The concentration of Bax protein in HIBD+UCBMC group was lower than that in the HIBD+NS group (P<0.01). The concentration of Bcl-2 protein was higher compared with the HIBD+NS, N+NS and N+UCBMC groups (P<0.05). CONCLUSIONS UCBMC transplantation via lateral ventricle can upregulate the expression of Bcl-2 protein and down-regulate the expression of Bax protein, thus alleviating brain neural apoptosis in neonatal rats with HIBD.
Collapse
|
35
|
Abstract
Cerebral palsy is the most common cause of childhood-onset, lifelong physical disability in most countries, affecting about 1 in 500 neonates with an estimated prevalence of 17 million people worldwide. Cerebral palsy is not a disease entity in the traditional sense but a clinical description of children who share features of a non-progressive brain injury or lesion acquired during the antenatal, perinatal or early postnatal period. The clinical manifestations of cerebral palsy vary greatly in the type of movement disorder, the degree of functional ability and limitation and the affected parts of the body. There is currently no cure, but progress is being made in both the prevention and the amelioration of the brain injury. For example, administration of magnesium sulfate during premature labour and cooling of high-risk infants can reduce the rate and severity of cerebral palsy. Although the disorder affects individuals throughout their lifetime, most cerebral palsy research efforts and management strategies currently focus on the needs of children. Clinical management of children with cerebral palsy is directed towards maximizing function and participation in activities and minimizing the effects of the factors that can make the condition worse, such as epilepsy, feeding challenges, hip dislocation and scoliosis. These management strategies include enhancing neurological function during early development; managing medical co-morbidities, weakness and hypertonia; using rehabilitation technologies to enhance motor function; and preventing secondary musculoskeletal problems. Meeting the needs of people with cerebral palsy in resource-poor settings is particularly challenging.
Collapse
|
36
|
Wu Q, Chen W, Sinha B, Tu Y, Manning S, Thomas N, Zhou S, Jiang H, Ma H, Kroessler DA, Yao J, Li Z, Inder TE, Wang X. Neuroprotective agents for neonatal hypoxic-ischemic brain injury. Drug Discov Today 2015; 20:1372-81. [PMID: 26360053 DOI: 10.1016/j.drudis.2015.09.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 07/31/2015] [Accepted: 09/01/2015] [Indexed: 01/13/2023]
Abstract
Hypoxic-ischemic (H-I) brain injury in newborns is a major cause of morbidity and mortality that claims thousands of lives each year. In this review, we summarize the promising neuroprotective agents tested on animal models and pilot clinical studies of neonatal H-I brain injury according to the different phases of the disease. These agents target various phases of injury including the early phase of excitotoxicity, oxidative stress and apoptosis as well as late-phase inflammatory reaction and neural repair. We analyze the cell survival and cell death pathways modified by these agents in neonatal H-I brain injury. We aim to 'build a bridge' between animal trials of neuroprotective agents and potential candidate treatments for future clinical applications against H-I encephalopathy.
Collapse
Affiliation(s)
- Qiaofeng Wu
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610091, China
| | - Wu Chen
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Clinical Laboratory, Dongfeng Hospital of Hubei University of Medicine, Shiyan, Hubei 442012, China
| | - Bharati Sinha
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Pediatrics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Yanyang Tu
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Simon Manning
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Niranjan Thomas
- Department of Neonatology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hong Jiang
- Department of Pediatrics, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China
| | - He Ma
- Third Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530031, China
| | - Daphne A Kroessler
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jiemin Yao
- Third Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530031, China
| | - Zhipu Li
- Department of Pediatrics, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China
| | - Terry E Inder
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
37
|
Chen LX, Ma SM, Zhang P, Fan ZC, Xiong M, Cheng GQ, Yang Y, Qiu ZL, Zhou WH, Li J. Neuroprotective effects of oligodendrocyte progenitor cell transplantation in premature rat brain following hypoxic-ischemic injury. PLoS One 2015; 10:e0115997. [PMID: 25790286 PMCID: PMC4366232 DOI: 10.1371/journal.pone.0115997] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/03/2014] [Indexed: 01/19/2023] Open
Abstract
Periventricular leukomalacia (PVL) is a common ischemic brain injury in premature infants for which there is no effective treatment. The objective of this study was to determine whether transplanted mouse oligodendrocyte progenitor cells (OPCs) have neuroprotective effects in a rat model of PVL. Hypoxia-ischemia (HI) was induced in 3-day-old rat pups by left carotid artery ligation, followed by exposure to 6% oxygen for 2.5 h. Animals were assigned to OPC transplantation or sham control groups and injected with OPCs or PBS, respectively, and sacrificed up to 6 weeks later for immunohistochemical analysis to investigate the survival and differentiation of transplanted OPCs. Apoptosis was evaluated by double immunolabeling of brain sections for caspase-3 and neuronal nuclei (NeuN), while proliferation was assessed using a combination of anti-Nestin and -bromodeoxyuridine antibodies. The expression of brain-derived neurotrophic factor (BDNF) and Bcl-2 was examined 7 days after OPC transplantation. The Morris water maze was used to test spatial learning and memory. The results showed that transplanted OPCs survived and formed a myelin sheath, and stimulated BDNF and Bcl-2 expression and the proliferation of neural stem cells (NSC), while inhibiting HI-induced neuronal apoptosis relative to control animals. Moreover, deficits in spatial learning and memory resulting from HI were improved by OPC transplantation. These results demonstrate an important neuroprotective role for OPCs that can potentially be exploited in cell-based therapeutic approaches to minimize HI-induced brain injury.
Collapse
Affiliation(s)
- Long-Xia Chen
- Key Laboratory of Birth Defect, Children’s Hospital of Fudan University, Shanghai, China
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
| | - Si-Min Ma
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, 201102, China
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
| | - Peng Zhang
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
| | - Zi-Chuan Fan
- Key Laboratory of Birth Defect, Children’s Hospital of Fudan University, Shanghai, China
| | - Man Xiong
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
| | - Guo-Qiang Cheng
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, 201102, China
| | - Yi Yang
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
| | - Zi-Long Qiu
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wen-Hao Zhou
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, 201102, China
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
- * E-mail: (WZ); (JL)
| | - Jin Li
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- * E-mail: (WZ); (JL)
| |
Collapse
|
38
|
Yang JJ, Tao H, Li J. Hedgehog signaling pathway as key player in liver fibrosis: new insights and perspectives. Expert Opin Ther Targets 2014; 18:1011-21. [PMID: 24935558 DOI: 10.1517/14728222.2014.927443] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Activation of hepatic stellate cells (HSCs) is a pivotal cellular event in liver fibrosis. Therefore, improving our understanding of the molecular pathways that are involved in these processes is essential to generate new therapies for liver fibrosis. Greater knowledge of the role of the hedgehog signaling pathway in liver fibrosis could improve understanding of the liver fibrosis pathogenesis. AREAS COVERED The aim of this review is to describe the present knowledge about the hedgehog signaling pathway, which significantly participates in liver fibrosis and HSC activation, and look ahead on new perspectives of hedgehog signaling pathway research. Moreover, we will discuss the different interactions with hedgehog signaling pathway-regulated liver fibrosis. EXPERT OPINION The hedgehog pathway modulates several important aspects of function, including cell proliferation, activation and differentiation. Targeting the hedgehog pathway can be a promising direction in liver fibrosis treatment. We discuss new perspectives of hedgehog signaling pathway activation in liver fibrosis and HSC fate, including DNA methylation, methyl CpG binding protein 2, microRNA, irradiation and metabolism that influence hedgehog signaling pathway transduction. These findings identify the hedgehog pathway as a potentially important for biomarker development and therapeutic targets in liver fibrosis. Future studies are needed in order to find safer and more effective hedgehog-based drugs.
Collapse
Affiliation(s)
- Jing-Jing Yang
- The Second Hospital of Anhui Medical University, Department of Pharmacology , Hefei 230601 , China
| | | | | |
Collapse
|
39
|
Mouhieddine TH, Kobeissy FH, Itani M, Nokkari A, Wang KK. Stem cells in neuroinjury and neurodegenerative disorders: challenges and future neurotherapeutic prospects. Neural Regen Res 2014; 9:901-6. [PMID: 25206908 PMCID: PMC4146225 DOI: 10.4103/1673-5374.133129] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2014] [Indexed: 12/15/2022] Open
Abstract
The prevalence of neurodegenerative diseases and neural injury disorders is increasing worldwide. Research is now focusing on improving current neurogenesis techniques including neural stem cell therapy and other biochemical drug-based approaches to ameliorate these disorders. Unfortunately, we are still facing many obstacles that are rendering current neurotherapies ineffective in clinical trials for reasons that are yet to be discovered. That is why we should start by fully understanding the complex mechanisms of neurogenesis and the factors that affect it, or else, all our suggested therapies would fail since they would not be targeting the essence of the neurological disorder but rather the symptoms. One possible paradigm shift is to switch from neuroprotectant therapies towards neurodegeneration/neurorestorative approaches. In addition, other and our laboratories are increasingly focusing on combining the use of pharmacological agents (such as Rho-associated kinase (ROCK) inhibitors or other growth factors (such as brain-derived neurotrophic factor (BDNF)) and stem cell treatment to enhance the survivability and/or differentiation capacity of transplanted stem cells in neurotrauma or other neurodegeneration animal models. Ongoing stem cell research is surely on the verge of a breakthrough of multiple effective therapeutic options for neurodegenerative disorders. Once, we fully comprehend the process of neurogenesis and its components, we will fully be capable of manipulating and utilizing it. In this work, we discuss the current knowledge of neuroregenerative therapies and their associated challenges.
Collapse
Affiliation(s)
- Tarek H. Mouhieddine
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Firas H. Kobeissy
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Center for Neuroproteomics and Biomarkers Research, Department of Psychiatry, Gainesville, Division of Addiction Medicine, Department of Psychiatry, University of Florida, Gainesville, FL 32610, USA
| | - Muhieddine Itani
- Faculty of Medicine, Saint George University of London, Nicosia, Cyprus
| | - Amaly Nokkari
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Kevin K.W. Wang
- Center for Neuroproteomics and Biomarkers Research, Department of Psychiatry, Gainesville, Division of Addiction Medicine, Department of Psychiatry, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
40
|
Wang X, Zhao Y, Wang X. Umbilical cord blood cells regulate the differentiation of endogenous neural stem cells in hypoxic ischemic neonatal rats via the hedgehog signaling pathway. Brain Res 2014; 1560:18-26. [PMID: 24565927 DOI: 10.1016/j.brainres.2014.02.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 01/31/2014] [Accepted: 02/08/2014] [Indexed: 10/25/2022]
Abstract
Transplantation of umbilical cord blood mononuclear cells (UCBMC) promotes the proliferation of endogenous neural stem cells (NSCs), but it has been unclear whether the proliferating NSCs can differentiate into mature neural cells. Therefore, we explored the effects of UCBMC transplantation on the differentiation of endogenous NSCs and their underlying mechanisms. Seven-day-old Sprague-Dawley rats underwent left carotid ligation followed by hypoxic stress. UCBMC were transplanted 24h after hypoxia ischemia (HI). BrdU/β-tubulin/HNA/DAPI, BrdU/GFAP/HNA/DAPI, Ngn1/DAPI, and BMP4/DAPI were measured by immunofluorescence staining; Shh, Gli1, Ngn1, and BMP4 proteins were measured by western-blot analysis 28 days after transplantation. More newborn neurons and fewer astrocytes were observed in the HI+UCBMC group, its neuronal percentage was higher, and glial percentage was lower compared with the N+UCBMC (P<0.05) and HI+PBS groups (P<0.01), while fewer newborn neurons and more newborn astrocytes were found in the HI+cyclopamine (an antagonist of the hedgehog protein)+UCBMC group compared with the HI+UCBMC group (P<0.01). The expression of Shh, Gli1, and Ngn1 proteins was higher and BMP4 protein was lower in the HI+UCBMC compared with the HI+PBS group (P<0.01) and the HI+cyclopamine+UCBMC group (P<0.01). Linear regression analysis showed that the differentiation of NSCs correlated with expression of Ngn1 and BMP4 proteins (P<0.01). In conclusion, UCBMC promote neuronal differentiation and reduce glial differentiation in hypoxic ischemic neonatal rats via the hedgehog signaling pathway.
Collapse
Affiliation(s)
- Xiaoli Wang
- Department of Medical Imaging, Weifang Medical University, Weifang, Shandong, China.
| | - Yansong Zhao
- Department of Ophthalmology, Weifang Medical University Affiliated Hospital, Clinical Institute, Weifang Medical University, Weifang, Shandong, China
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women׳s Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
41
|
Effects of intravenous administration of umbilical cord blood CD34(+) cells in a mouse model of neonatal stroke. Neuroscience 2014; 263:148-58. [PMID: 24444827 DOI: 10.1016/j.neuroscience.2014.01.018] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 01/06/2014] [Accepted: 01/06/2014] [Indexed: 12/16/2022]
Abstract
Neonatal stroke occurs in approximately 1/4000 live births and results in life-long neurological impairments: e.g., cerebral palsy. Currently, there is no evidence-based specific treatment for neonates with stroke. Several studies have reported the benefits of umbilical cord blood (UCB) cell treatment in rodent models of neonatal brain injury. However, all of the studies examined the effects of administering either the UCB mononuclear cell fraction or UCB-derived mesenchymal stem cells in neonatal rat models. The objective of this study was to examine the effects of human UCB CD34(+) cells (hematopoietic stem cell/endothelial progenitor cells) in a mouse model of neonatal stroke, which we recently developed. On postnatal day 12, immunocompromized (SCID) mice underwent permanent occlusion of the left middle cerebral artery (MCAO). Forty-eight hours after MCAO, human UCB CD34(+) cells (1×10(5)cells) were injected intravenously into the mice. The area in which cerebral blood flow (CBF) was maintained was temporarily larger in the cell-treated group than in the phosphate-buffered saline (PBS)-treated group at 24h after treatment. With cell treatment, the percent loss of ipsilateral hemispheric volume was significantly ameliorated (21.5±1.9%) compared with the PBS group (25.6±5.1%) when assessed at 7weeks after MCAO. The cell-treated group did not exhibit significant differences from the PBS group in either rotarod (238±46s in the sham-surgery group, 175±49s in the PBS group, 203±54s in the cell-treated group) or open-field tests. The intravenous administration of human UCB CD34(+) cells modestly reduced histological ischemic brain damage after neonatal stroke in mice, with a transient augmentation of CBF in the peri-infarct area.
Collapse
|
42
|
Castillo-Melendez M, Yawno T, Jenkin G, Miller SL. Stem cell therapy to protect and repair the developing brain: a review of mechanisms of action of cord blood and amnion epithelial derived cells. Front Neurosci 2013; 7:194. [PMID: 24167471 PMCID: PMC3807037 DOI: 10.3389/fnins.2013.00194] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 10/07/2013] [Indexed: 12/12/2022] Open
Abstract
In the research, clinical, and wider community there is great interest in the use of stem cells to reduce the progression, or indeed repair brain injury. Perinatal brain injury may result from acute or chronic insults sustained during fetal development, during the process of birth, or in the newborn period. The most readily identifiable outcome of perinatal brain injury is cerebral palsy, however, this is just one consequence in a spectrum of mild to severe neurological deficits. As we review, there are now clinical trials taking place worldwide targeting cerebral palsy with stem cell therapies. It will likely be many years before strong evidence-based results emerge from these trials. With such trials underway, it is both appropriate and timely to address the physiological basis for the efficacy of stem-like cells in preventing damage to, or regenerating, the newborn brain. Appropriate experimental animal models are best placed to deliver this information. Cell availability, the potential for immunological rejection, ethical, and logistical considerations, together with the propensity for native cells to form teratomas, make it unlikely that embryonic or fetal stem cells will be practical. Fortunately, these issues do not pertain to the use of human amnion epithelial cells (hAECs), or umbilical cord blood (UCB) stem cells that are readily and economically obtained from the placenta and umbilical cord discarded at birth. These cells have the potential for transplantation to the newborn where brain injury is diagnosed or even suspected. We will explore the novel characteristics of hAECs and undifferentiated UCB cells, as well as UCB-derived endothelial progenitor cells (EPCs) and mesenchymal stem cells (MSCs), and how immunomodulation and anti-inflammatory properties are principal mechanisms of action that are common to these cells, and which in turn may ameliorate the cerebral hypoxia and inflammation that are final pathways in the pathogenesis of perinatal brain injury.
Collapse
Affiliation(s)
- Margie Castillo-Melendez
- The Ritchie Centre, Monash Institute of Medical Research, Monash University Clayton, VIC, Australia
| | | | | | | |
Collapse
|