1
|
Michór P, Renardson L, Li S, Boltze J. Neurorestorative Approaches for Ischemic StrokeChallenges, Opportunities, and Recent Advances. Neuroscience 2024; 550:69-78. [PMID: 38763225 DOI: 10.1016/j.neuroscience.2024.05.009] [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: 11/11/2023] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/21/2024]
Abstract
Despite recent advances in acute stroke management, most patients experiencing a stroke will suffer from residual brain damage and functional impairment. Addressing those residual deficits would require neurorestoration, i.e., rebuilding brain tissue to repair the structural brain damage caused by stroke. However, there are major pathobiological, anatomical and technological hurdles making neurorestorative approaches remarkably challenging, and true neurorestoration after larger ischemic lesions could not yet be achieved. On the other hand, there has been steady advancement in our understanding of the limits of tissue regeneration in the adult mammalian brain as well as of the fundamental organization of brain tissue growth during embryo- and ontogenesis. This has been paralleled by the development of novel animal models to study stroke, advancement of biomaterials that can be used to support neurorestoration, and in stem cell technologies. This review gives a detailed explanation of the major hurdles so far preventing the achievement of neurorestoration after stroke. It will also describe novel concepts and advancements in biomaterial science, brain organoid culturing, and animal modeling that may enable the investigation of post-stroke neurorestorative approaches in translationally relevant setups. Finally, there will be a review of recent achievements in experimental studies that have the potential to be the starting point of research and development activities that may eventually bring post-stroke neurorestoration within reach.
Collapse
Affiliation(s)
- Paulina Michór
- University of Warwick, School of Life Sciences, Coventry CV4 7AL, United Kingdom
| | - Lydia Renardson
- University of Warwick, Warwick Medical School, Coventry CV4 7AL, United Kingdom
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Johannes Boltze
- University of Warwick, School of Life Sciences, Coventry CV4 7AL, United Kingdom.
| |
Collapse
|
2
|
Detante O, Legris L, Moisan A, Rome C. Cell Therapy and Functional Recovery of Stroke. Neuroscience 2024; 550:79-88. [PMID: 38013148 DOI: 10.1016/j.neuroscience.2023.11.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 11/29/2023]
Abstract
Stroke is the most common cause of disability. Brain repair mechanisms are often insufficient to allow a full recovery. Stroke damage involve all brain cell type and extracellular matrix which represent the crucial "glio-neurovascular niche" useful for brain plasticity. Regenerative medicine including cell therapies hold great promise to decrease post-stroke disability of many patients, by promoting both neuroprotection and neural repair through direct effects on brain lesion and/or systemic effects such as immunomodulation. Mechanisms of action vary according to each grafted cell type: "peripheral" stem cells, such as mesenchymal stem cells (MSC), can provide paracrine trophic support, and neural stem/progenitor cells (NSC) or neurons can act as direct cells' replacements. Optimal time window, route, and doses are still debated, and may depend on the chosen medicinal product and its expected mechanism such as neuroprotection, delayed brain repair, systemic effects, or graft survival and integration in host network. MSC, mononuclear cells (MNC), umbilical cord stem cells and NSC are the most investigated. Innovative approaches are implemented concerning combinatorial approaches with growth factors and biomaterials such as injectable hydrogels which could protect a cell graft and/or deliver drugs into the post-stroke cavity at chronic stages. Through main publications of the last two decades, we provide in this review concepts and suggestions to improve future translational researches and larger clinical trials of cell therapy in stroke.
Collapse
Affiliation(s)
- Olivier Detante
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institute Neurosciences, 38000 Grenoble, France; Stroke Unit, Neurology, CHU Grenoble Alpes, CS10217, 38043 Grenoble, France; Axe Neurosciences Cliniques - Innovative Brain Therapies, CHU Grenoble Alpes, 38000 Grenoble, France.
| | - Loic Legris
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institute Neurosciences, 38000 Grenoble, France; Stroke Unit, Neurology, CHU Grenoble Alpes, CS10217, 38043 Grenoble, France; Axe Neurosciences Cliniques - Innovative Brain Therapies, CHU Grenoble Alpes, 38000 Grenoble, France.
| | - Anaick Moisan
- Axe Neurosciences Cliniques - Innovative Brain Therapies, CHU Grenoble Alpes, 38000 Grenoble, France; Cell Therapy and Engineering Unit, EFS Rhône Alpes, 464 route de Lancey, 38330 Saint Ismier, France.
| | - Claire Rome
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institute Neurosciences, 38000 Grenoble, France; Stroke Unit, Neurology, CHU Grenoble Alpes, CS10217, 38043 Grenoble, France; Axe Neurosciences Cliniques - Innovative Brain Therapies, CHU Grenoble Alpes, 38000 Grenoble, France.
| |
Collapse
|
3
|
Whitelaw A, Thoresen M. Therapeutic Hypothermia for Hypoxic-Ischemic Brain Injury Is More Effective in Newborn Infants than in Older Patients: Review and Hypotheses. Ther Hypothermia Temp Manag 2023; 13:170-174. [PMID: 37638830 DOI: 10.1089/ther.2023.0050] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023] Open
Abstract
Posthypoxic therapeutic hypothermia has been tested in newborn infants, with seven randomized trials showing consistent evidence of reduction in death, cerebral palsy, and cognitive impairment at school age. In contrast, randomized trials of hypothermia after cardiac arrest in adults have not shown consistent evidence of lasting neurological protection. The apparently greater effectiveness of therapeutic hypothermia in newborns may be due to important biological and clinical differences. One such difference is that adults are heavily colonized with microbes, and many have active inflammatory processes at the time of arrest, but few newborns are heavily colonized or infected at the time of birth. Inflammation can interfere with hypothermia's neuroprotection. A second difference is that apoptosis is more commonly the pathway of neuronal death in newborns than in adults. Hypothermia inhibits apoptosis but not necrosis. Newborns have a larger endogenous supply of stem cells (which reduce apoptosis) than adults and this may favor regeneration and protection from hypothermia and regeneration. A third difference is that immature oligodendroglia are more sensitive to free radical attack then mature oligodendroglia. Hypothermia reduces free radical release. In addition, immature brain has increased N-methyl-D-aspartate receptor subunits compared with adults and hypothermia reduces excitotoxic amino acids. Adults suffering cardiac arrest often have comorbidities such as diabetes, hypertension, and atherosclerosis, which complicate recovery, but newborn infants rarely have comorbidities before asphyxia. Adult hypothermia treatment may have been too short as no trial has cooled for longer than 48 hours, some only 24 or 12 hours, but neonatal therapeutic hypothermia has routinely lasted 72 hours. We hypothesize that this combination of differences favors the effectiveness of therapeutic hypothermia in newborn infants compared with adults.
Collapse
Affiliation(s)
- Andrew Whitelaw
- Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Marianne Thoresen
- Translational Health Sciences, University of Bristol, Bristol, United Kingdom
- Department of Physiology, Institute of Basic Medical Research, University of Oslo, Oslo, Norway
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Cox CS, Juranek J, Kosmach S, Pedroza C, Thakur N, Dempsey A, Rennie K, Scott MC, Jackson M, Kumar A, Aertker B, Caplan H, Triolo F, Savitz SI. Autologous cellular therapy for cerebral palsy: a randomized, crossover trial. Brain Commun 2022; 4:fcac131. [PMID: 35702731 PMCID: PMC9188321 DOI: 10.1093/braincomms/fcac131] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/24/2022] [Accepted: 05/17/2022] [Indexed: 11/14/2022] Open
Abstract
We examined an autologous mononuclear-cell-therapy-based approach to treat cerebral palsy using autologous umbilical cord blood or bone-marrow-derived mononuclear cells. The primary objective was to determine if autologous cells are safe to administer in children with cerebral palsy. The secondary objectives were to determine if there was improvement in motor function of patients 12 months after infusion using the Gross Motor Function Measure and to evaluate impact of treatment on corticospinal tract microstructure as determined by radial diffusivity measurement. This Phase 1/2a trial was a randomized, blinded, placebo-controlled, crossover study in children aged 2-10 years of age with cerebral palsy enrolled between November 2013 and November 2016. Participants were randomized to 2:1 treatment:placebo. Treatment was either autologous bone-marrow-derived mononuclear cells or autologous umbilical cord blood. All participants who enrolled and completed their baseline visit planned to return for follow-up visits at 6 months, 12 months and 24 months after the baseline visit. At the 12-month post-treatment visit, participants who originally received the placebo received either bone-marrow-derived mononuclear cell or umbilical cord blood treatment. Twenty participants were included; 7 initially randomized to placebo, and 13 randomized to treatment. Five participants randomized to placebo received bone-marrow-derived mononuclear cells, and 2 received umbilical cord blood at the 12-month visit. None of the participants experienced adverse events related to the stem cell infusion. Cell infusion at the doses used in our study did not dramatically alter motor function. We observed concordant bilateral changes in radial diffusivity in 10 of 15 cases where each corticospinal tract could be reconstructed in each hemisphere. In 60% of these cases (6/10), concordant decreases in bilateral corticospinal tract radial diffusivity occurred post-treatment. In addition, 100% of unilateral corticospinal tract cases (3/3) exhibited decreased corticospinal tract radial diffusivity post-treatment. In our discordant cases (n = 5), directionality of changes in corticospinal tract radial diffusivity appeared to coincide with handedness. There was a significant improvement in corticospinal tract radial diffusivity that appears related to handedness. Connectivity strength increased in either or both pathways (corticio-striatal and thalamo-cortical) in each participant at 12 months post-treatment. These data suggest that both stem cell infusions are safe. There may be an improvement in myelination in some groups of patients that correlate with small improvements in the Gross Motor Function Measure scales. A larger autologous cord blood trial is impractical at current rates of blood banking. Either increased private banking or matched units would be required to perform a larger-scale trial.
Collapse
Affiliation(s)
- Charles S. Cox
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Program in Pediatric Regenerative Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Jenifer Juranek
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Program in Pediatric Regenerative Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Steven Kosmach
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Claudia Pedroza
- Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Nivedita Thakur
- Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Allison Dempsey
- Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Kimberly Rennie
- Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Department of Neuropsychology, NeuroBehavioral Health, Milwaukee, WI, USA
| | - Michael C. Scott
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Margaret Jackson
- Department of Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Akshita Kumar
- Department of Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Benjamin Aertker
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Henry Caplan
- Department of Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Fabio Triolo
- Department of Pediatric Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Program in Pediatric Regenerative Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Sean I. Savitz
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| |
Collapse
|
7
|
Jin X, Li P, Michalski D, Li S, Zhang Y, Jolkkonen J, Cui L, Didwischus N, Xuan W, Boltze J. Perioperative stroke: A perspective on challenges and opportunities for experimental treatment and diagnostic strategies. CNS Neurosci Ther 2022; 28:497-509. [PMID: 35224865 PMCID: PMC8928912 DOI: 10.1111/cns.13816] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 02/06/2023] Open
Abstract
Perioperative stroke is an ischemic or hemorrhagic cerebral event during or up to 30 days after surgery. It is a feared condition due to a relatively high incidence, difficulties in timely detection, and unfavorable outcome compared to spontaneously occurring stroke. Recent preclinical data suggest that specific pathophysiological mechanisms such as aggravated neuroinflammation contribute to the detrimental impact of perioperative stroke. Conventional treatment options are limited in the perioperative setting due to difficult diagnosis and medications affecting coagulation in may cases. On the contrary, the chance to anticipate cerebrovascular events at the time of surgery may pave the way for prevention strategies. This review provides an overview on perioperative stroke incidence, related problems, and underlying pathophysiological mechanisms. Based on this analysis, we assess experimental stroke treatments including neuroprotective approaches, cell therapies, and conditioning medicine strategies regarding their potential use in perioperative stroke. Interestingly, the specific aspects of perioperative stroke might enable a more effective application of experimental treatment strategies such as classical neuroprotection whereas others including cell therapies may be of limited use. We also discuss experimental diagnostic options for perioperative stroke augmenting classical clinical and imaging stroke diagnosis. While some experimental stroke treatments may have specific advantages in perioperative stroke, the paucity of established guidelines or multicenter clinical research initiatives currently limits their thorough investigation.
Collapse
Affiliation(s)
- Xia Jin
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | | | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yueman Zhang
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Jukka Jolkkonen
- Department of Neurology and A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Lili Cui
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Nadine Didwischus
- School of Life Sciences, University of Warwick, Coventry, UK.,Department of Radiology, University of Pittsburgh, Pittsburgh, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Wei Xuan
- Department of Anesthesiology, Renji Hospital, School of Medicine Shanghai Jiaotong University, Shanghai, China
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, UK
| |
Collapse
|
8
|
Satani N, Parsha K, Savitz SI. Enhancing Stroke Recovery With Cellular Therapies. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00062-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
9
|
Lee TK, Lu CY, Tsai ST, Tseng PH, Lin YC, Lin SZ, Wang JC, Huang CY, Chiu TL. Complete Restoration of Motor Function in Acute Cerebral Stroke Treated with Allogeneic Human Umbilical Cord Blood Monocytes: Preliminary Results of a phase I Clinical Trial. Cell Transplant 2021; 30:9636897211067447. [PMID: 34939863 PMCID: PMC8728774 DOI: 10.1177/09636897211067447] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Stem cell therapy has been explored for the treatment of cerebral stroke. Several types of stem cells have been investigated to ensure the safety and efficacy in clinical trials.Cryopreserved umbilical cord blood (UCB) mononuclear cells (MNCs) obtained from healthy donors have a more stabilized quality, thereby ensuring a successful therapy. A phase I study was conducted on patients aged 45-80 years who sustained acute ischemic stroke. An UCB unit was obtained from a public cord blood bank based on ABO/Rh blood type, HLA matching score (6/6), and cell dose (total MNC count of 0.5-5 × 107 cells/kg). In addition, to facilitate blood brain barrier penetration of UCB, 4 doses of 100 mL mannitol was administered intravenously after 30 min after UCB transplantation and every 4 h thereafter. The primary outcomes were the number of disease (GVHD) within 100 days after transfusion. The secondary outcomes were changes in the National Institutes of Health Stroke Scale (NIHSS), Barthel index, and Berg Balance Scale scores. A 46-year-old male patient with identical ABO/Rh blood type, HLA matching score of 6/6, and MNC count of 2.63 × 108 cells/kg was enrolled. The patient did not present with serious AEs or GVHD during the 12-month study period. The patient's NIHSS score decreased from 9 to 1. Moreover, the Berg Balance Scale score increased from 0 to 48 and the Barthel index score from 0 to 90. This preliminary study showed that an adult patient with hemiplegia due to ischemic stroke completely recovered within 12 months after receiving allogeneic UCB therapy.
Collapse
Affiliation(s)
- Tian-Kuo Lee
- Department of Neurosurgical Unit, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien
| | - Cheng-You Lu
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien
| | - Sheng-Tzung Tsai
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien.,College of Medicine, Tzu Chi University, Hualien
| | - Pao-Hui Tseng
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien.,College of Medicine, Tzu Chi University, Hualien
| | - Yu-Chen Lin
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien
| | - Shinn-Zong Lin
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien.,College of Medicine, Tzu Chi University, Hualien.,Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien
| | | | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien.,Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung.,Graduate Institute of Biomedical Science, China Medical University, Taichung.,Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien.,Department of Medical Research, China Medical University, Hospital, China Medical University, Taichung
| | - Tsung-Lang Chiu
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien.,College of Medicine, Tzu Chi University, Hualien
| |
Collapse
|
10
|
Mangin G, Kubis N. Cell Therapy for Ischemic Stroke: How to Turn a Promising Preclinical Research into a Successful Clinical Story. Stem Cell Rev Rep 2020; 15:176-193. [PMID: 30443706 DOI: 10.1007/s12015-018-9864-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Stroke is a major public health issue with limited treatment. The pharmacologically or mechanically removing of the clot is accessible to less than 10% of the patients. Stem cell therapy is a promising alternative strategy since it increases the therapeutic time window but many issues remain unsolved. To avoid a new dramatic failure when translating experimental data on the bedside, this review aims to highlight the indispensable checkpoints to make a successful clinical trial based on the current preclinical literature. The large panel of progenitors/ stem cells at the researcher's disposal is to be used wisely, regarding the type of cells, the source of cells, the route of delivery, the time window, since it will directly affect the outcome. Mechanisms are still incompletely understood, although recent studies have focused on the inflammation modulation of most cells types.
Collapse
Affiliation(s)
| | - Nathalie Kubis
- INSERM U965, F-75475, Paris, France. .,Sorbonne Paris Cité, Université Paris Diderot, F-75475, Paris, France. .,Service de Physiologie Clinique-Explorations Fonctionnelles, AP-HP, Hôpital Lariboisière, 2 rue Ambroise Paré, F-75475, Paris, France.
| |
Collapse
|
11
|
Lin KC, Chai HT, Chen KH, Sung PH, Chiang JY, Shao PL, Huang CR, Li YC, Ko SF, Yip HK. Intra-carotid arterial transfusion of circulatory-derived autologous endothelial progenitor cells in rodent after ischemic stroke-evaluating the impact of therapeutic time points on prognostic outcomes. Stem Cell Res Ther 2020; 11:219. [PMID: 32503671 PMCID: PMC7275327 DOI: 10.1186/s13287-020-01739-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/06/2020] [Accepted: 05/20/2020] [Indexed: 12/25/2022] Open
Abstract
Background This study tested the optimal time point for left intra-carotid arterial (LICA) administration of circulatory-derived autologous endothelial progenitor cells (EPCs) for improving the outcome in rat after acute ischemic stroke (IS). Methods and results Adult male SD rats (n = 70) were equally categorized into group 1 (sham-operated control), group 2 (IS), group 3 (IS+EPCs/1.2 × 106 cells/by LICA administration 3 h after IS), group 4 (IS+EPCs/LICA administration post-day-3 IS), group 5 (IS+EPCs/LICA administration post-day-7 IS), group 6 (IS+EPCs/LICA administration post-day-14 IS), and group 7 (IS+EPCs/LICA administration post-day-28 IS). The brain infarct volume (BIV) (at day 60/MRI) was lowest in group 1, highest in group 2, and significantly progressively increased from groups 3 to 7, whereas among the IS animals, the neurological function was significantly preserved in groups 3 to 6 than in groups 2 and 7 post-day-60 IS (all P < 0.0001). By day 60, the endothelial cell markers at protein and cellular levels and number of small vessels exhibited an opposite pattern of BIV among the groups (all P < 0.0001). The protein and cellular levels of inflammation, and protein levels of oxidative stress, autophagy, and apoptosis were highest in group 2, lowest in group 1, and progressively increased from groups 3 to 7 (all P < 0.0001). The angiogenesis biomarkers at protein and cellular levels were significantly progressively increased from groups 1 to 3, then significantly progressively decreased from groups 4 to 7 (all P < 0.0001). Conclusion Early EPC administration provided better benefits on improving functional/image/molecular-cellular outcomes after acute IS in rat.
Collapse
Affiliation(s)
- Kun-Chen Lin
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Han-Tan Chai
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung, Taiwan
| | - Kuan-Hung Chen
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Pei-Hsun Sung
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung, Taiwan.,Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - John Y Chiang
- Department of Computer Science and Engineering, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Pei-Lin Shao
- Department of Nursing, Asia University, Taichung, Taiwan
| | - Chi-Ruei Huang
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung, Taiwan.,Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yi-Chen Li
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung, Taiwan
| | - Sheung-Fat Ko
- Department of Radiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hon-Kan Yip
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung, Taiwan. .,Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan. .,Department of Nursing, Asia University, Taichung, Taiwan. .,Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan. .,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan. .,Division of Cardiology, Department of Internal Medicine, Xiamen Chang Gung Hospital, Xiamen, Fujian, China.
| |
Collapse
|
12
|
Boltze J, Abe K, Clarkson AN, Detante O, Pimentel-Coelho PM, Rosado-de-Castro PH, Janowski M. Editorial: Cell-based Therapies for Stroke: Promising Solution or Dead End? Front Neurol 2020; 11:171. [PMID: 32308639 PMCID: PMC7145965 DOI: 10.3389/fneur.2020.00171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 02/24/2020] [Indexed: 11/13/2022] Open
Affiliation(s)
- Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Koji Abe
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Andrew N Clarkson
- Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Oliver Detante
- Stroke Unit, Neurology Department, Grenoble Hospital, Grenoble, France.,Grenoble Institute of Neurosciences, Inserm U1216, Université Grenoble Alpes, Grenoble, France
| | - Pedro M Pimentel-Coelho
- Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil
| | - Paulo H Rosado-de-Castro
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Radiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,D'Or Institute for Research and Education, Rio de Janeiro, Brazil
| | - Miroslaw Janowski
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, United States.,NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
13
|
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: 1.8] [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
|
14
|
Kamarudin SN, Iezhitsa I, Tripathy M, Alyautdin R, Ismail NM. Neuroprotective effect of poly(lactic-co-glycolic acid) nanoparticle-bound brain-derived neurotrophic factor in a permanent middle cerebral artery occlusion model of ischemia in rats. Acta Neurobiol Exp (Wars) 2020. [DOI: 10.21307/ane-2020-001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
15
|
Boltze J, Jolkkonen J. Safety evaluation of intra-arterial cell delivery in stroke patients-a framework for future trials. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S271. [PMID: 32015990 DOI: 10.21037/atm.2019.12.07] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Jukka Jolkkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
16
|
Li J, Liu W, Yao W. Immortalized Human Bone Marrow Derived Stromal Cells in Treatment of Transient Cerebral Ischemia in Rats. J Alzheimers Dis 2019; 69:871-880. [PMID: 31156178 DOI: 10.3233/jad-190279] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Jianyuan Li
- Neurosurgical Department, The Affiliated Hospital of Qingdao University, Qingdao, China
- Neurosurgical Department, Rizhao City Hospital of Traditional Chinese Medicine, Rizhao, China
| | - Weidong Liu
- Neurosurgical Department, Liaocheng People’s Hospital, Liaocheng, China
| | - Weicheng Yao
- Neurosurgical Department, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
17
|
Cui LL, Golubczyk D, Tolppanen AM, Boltze J, Jolkkonen J. Cell therapy for ischemic stroke: Are differences in preclinical and clinical study design responsible for the translational loss of efficacy? Ann Neurol 2019; 86:5-16. [PMID: 31020699 DOI: 10.1002/ana.25493] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 04/01/2019] [Accepted: 04/21/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Li-Li Cui
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland
| | - Dominika Golubczyk
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | | | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Jukka Jolkkonen
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland.,Neurocenter, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
18
|
The Promising Effects of Transplanted Umbilical Cord Mesenchymal Stem Cells on the Treatment in Traumatic Brain Injury. J Craniofac Surg 2018; 29:1689-1692. [PMID: 30234712 PMCID: PMC6200375 DOI: 10.1097/scs.0000000000005042] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Many studies have reported the recovery ability of umbilical cord-derived mesenchymal stem cells (UC-MSCs) for neural diseases. In this study, the authors explored the roles of UC-MSCs to treat the traumatic brain injury. Umbilical cord-derived mesenchymal stem cells were isolated from healthy neonatal rat umbilical cord immediately after delivery. The traumatic brain injury (TBI) model was formed by the classical gravity method. The authors detected the behavior changes and measured the levels of inflammatory factors, such as interleukin-lβ and tumor necrosis factor-α by enzyme linked immunosorbent assay (ELISA) at 1, 2, 3, 4 weeks after transplantation between TBI treated and untreated with UC-MSCs. Simultaneously, the expression of glial cell line-derived neurotrophic factor (GDNF) and brain derived neurotrophic factor (BDNF) were measured by real-time-polymerase chain reaction and ELISA.The authors found that the group of transplantation UC-MSCs has a significant improvement than other group treated by phosphate buffered saline. In the behavioral test, the Neurological Severity Scores of UC-MSCs + TBI group were lower than TBI group (P < 0.05), but not obviously higher than control group at 2, 3, and 4week, respectively. The inflammatory factors are significantly reduced comparison with TBI group (P < 0.05), but both GDNF and BDNF were higher than TBI group (P < 0.05). The results indicated that UC-MSCs might play an important role in TBI recovery through inhibiting the release of inflammatory factors and increasing the expression of GDNF and BDNF.
Collapse
|
19
|
Lin W, Hsuan YCY, Lin MT, Kuo TW, Lin CH, Su YC, Niu KC, Chang CP, Lin HJ. Human Umbilical Cord Mesenchymal Stem Cells Preserve Adult Newborn Neurons and Reduce Neurological Injury after Cerebral Ischemia by Reducing the Number of Hypertrophic Microglia/Macrophages. Cell Transplant 2018; 26:1798-1810. [PMID: 29338384 PMCID: PMC5784525 DOI: 10.1177/0963689717728936] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Microglia are the first source of a neuroinflammatory cascade, which seems to be involved in every phase of stroke-related neuronal damage. Two weeks after transient middle cerebral artery occlusion (MCAO), vehicle-treated rats displayed higher numbers of total ionized calcium-binding adaptor molecule 1 (Iba-1)-positive cells, greater cell body areas of Iba-1-positive cells, and higher numbers of hypertrophic Iba-1-positive cells (with a cell body area over 80 μm2) in the ipsilateral ischemic brain regions including the frontal cortex, striatum, and parietal cortex. In addition, MCAO decreased the number of migrating neuroblasts (or DCX- and 5-ethynyl-2′-deoxyuridine-positive cells) in the cortex, subventricular zone, and hippocampus of the ischemic brain, followed by neurological injury (including brain infarct and neurological deficits). Intravenous administration of human umbilical cord–derived mesenchymal stem cells (hUC-MSCs; 1 × 106 or 4 × 106) at 24 h after MCAO reduced neurological injury, decreased the number of hypertrophic microglia/macrophages, and increased the number of newborn neurons in rat brains. Thus, the accumulation of hypertrophic microglia/macrophages seems to be detrimental to neurogenesis after stroke. Treatment with hUC-MSCs preserved adult newborn neurons and reduced functional impairment after transient cerebral ischemia by reducing the number of hypertrophic microglia/macrophages.
Collapse
Affiliation(s)
- Willie Lin
- 1 Meridigen Biotech Co., Ltd., Neihu, Taipei City, Taiwan
| | | | - Mao-Tsun Lin
- 2 Department of Medical Research, Chi Mei Medical Center, Tainan City, Taiwan
| | - Ting-Wei Kuo
- 3 Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan City, Taiwan
| | | | - Yu-Chin Su
- 1 Meridigen Biotech Co., Ltd., Neihu, Taipei City, Taiwan
| | - Ko-Chi Niu
- 4 Department of Hyperbaric Oxygen, Chi Mei Medical Center, Tainan City, Taiwan
| | - Ching-Ping Chang
- 2 Department of Medical Research, Chi Mei Medical Center, Tainan City, Taiwan.,3 Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan City, Taiwan.,5 The Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taipei City, Taiwan
| | - Hung-Jung Lin
- 3 Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan City, Taiwan.,6 Department of Emergency Medicine, Chi Mei Medical Center, Tainan City, Taiwan
| |
Collapse
|
20
|
Ehrhart J, Sanberg PR, Garbuzova-Davis S. Plasma derived from human umbilical cord blood: Potential cell-additive or cell-substitute therapeutic for neurodegenerative diseases. J Cell Mol Med 2018; 22:6157-6166. [PMID: 30334335 PMCID: PMC6237605 DOI: 10.1111/jcmm.13898] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022] Open
Abstract
Limited efficacy of current therapeutic approaches for neurodegenerative disease has led to increased interest in alternative therapies. Cord blood plasma (CBP) derived from human umbilical cord blood (hUCB) may be a potential therapeutic. Benefits of CBP injection into rodent models of aging or ischaemic stroke have been demonstrated, though how benefits are elicited is still unclear. The present study evaluated various factors within the same samples of CBP and human adult blood plasma/sera (ABP/S). Also, autologous CBP effects vs. ABP/S or foetal bovine serum supplements on mononuclear cells from hUCB (MNC hUCB) in vitro were determined. Results showed significantly low concentrations of pro-inflammatory cytokines (IL-2, IL-6, IFN-γ, and TNF-α) and elevated chemokine IL-8 in CBP. Significantly higher levels of VEGF, G-CSF, EGF and FGF-basic growth factors were determined in CBP vs. ABP/S. Autologous CBP media supplements significantly increased MNC hUCB viability and decreased apoptotic cell activity. We are first to demonstrate the unique CBP composition of cytokines and growth factors within the same CBP samples derived from hUCB. Also, our novel finding that autologous CBP promoted MNC hUCB viability and reduced apoptotic cell death in vitro supports CBP's potential as a sole therapeutic or cell-additive agent in developing therapies for various neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Paul R Sanberg
- Center of Excellence for Aging and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Psychiatry, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Svitlana Garbuzova-Davis
- Center of Excellence for Aging and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, Florida.,Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| |
Collapse
|
21
|
Abstract
Stroke still represents one of the most common causes of death and disability worldwide. Acute ischemic stroke (AIS), caused by brain arterial occlusion resulting from a thrombus or embolus, is the most common form of stroke. However, current therapies in AIS are inadequate, and the only US FDA approved treatment is the thrombolytic drug Alteplase. Therefore, establishing effective therapeutic strategies for AIS is urgently needed. Using nanoparticle-based technologies to deliver neuroprotective agents to the ischemic area has attracted increasing attention of late. In this review, the important molecular pathological mechanisms in cerebral ischemia are briefly summarized, the potential of nanoparticulate drug-delivery systems for AIS intervention and recovery are introduced and problems in the medical application of nanoparticles will also be discussed.
Collapse
|
22
|
Boshuizen MCS, Steinberg GK. Stem Cell-Based Immunomodulation After Stroke: Effects on Brain Repair Processes. Stroke 2018; 49:1563-1570. [PMID: 29724892 DOI: 10.1161/strokeaha.117.020465] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/05/2018] [Accepted: 03/20/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Marieke C S Boshuizen
- From the Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, CA
| | - Gary K Steinberg
- From the Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, CA.
| |
Collapse
|
23
|
Diabetic Ephrin-B2-Stimulated Peripheral Blood Mononuclear Cells Enhance Poststroke Recovery in Mice. Stem Cells Int 2018; 2018:2431567. [PMID: 29736174 PMCID: PMC5875038 DOI: 10.1155/2018/2431567] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/21/2017] [Accepted: 01/11/2018] [Indexed: 11/18/2022] Open
Abstract
Clinical trials of cell therapy in stroke favor autologous cell transplantation. To date, feasibility studies have used bone marrow-derived mononuclear cells, but harvesting bone marrow cells is invasive thus complicating bedside treatment. We investigated the therapeutic potential of peripheral blood-derived mononuclear cells (PB-MNC) harvested from diabetic patients and stimulated by ephrin-B2 (PB-MNC+) (500,000 cells), injected intravenously 18–24 hours after induced cerebral ischemia in mice. Infarct volume, neurological deficit, neurogenesis, angiogenesis, and inflammation were investigated as were the potential mechanisms of PB-MNC+ cells in poststroke neurorepair. At D3, infarct volume was reduced by 60% and 49% compared to unstimulated PB-MNC and PBS-treated mice, respectively. Compared to PBS, injection of PB-MNC+ increased cell proliferation in the peri-infarct area and the subventricular zone, decreased microglia/macrophage cell density, and upregulated TGF-β expression. At D14, microvessel density was decreased and functional recovery was enhanced compared to PBS-treated mice, whereas plasma levels of BDNF, a major regulator of neuroplasticity, were increased in mice treated with PB-MNC+ compared to the other two groups. Cell transcriptional analysis showed that ephrin-B2 induced phenotype switching of PB-MNC by upregulating genes controlling cell proliferation, inflammation, and angiogenesis, as confirmed by adhesion and Matrigel assays. Conclusions. This feasibility study suggests that PB-MNC+ transplantation poststroke could be a promising approach but warrants further investigation. If confirmed, this rapid, noninvasive bedside cell therapy strategy could be applied to stroke patients at the acute phase.
Collapse
|
24
|
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: 2.2] [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
|
25
|
Lin W, Hsuan YCY, Su YC, Lin CH, Lin MT, Chen ZH, Chang CP, Lin KC. CD34 - human placenta-derived mesenchymal stem cells protect against heat stroke mortality in rats. Oncotarget 2017; 9:1992-2001. [PMID: 29416747 PMCID: PMC5788615 DOI: 10.18632/oncotarget.23324] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/09/2017] [Indexed: 01/01/2023] Open
Abstract
CD34 is a transmembrane phosphoglycoprotein used to selectively enrich bone marrow in hematopoietic stem cells for transplantation. Treating rats with CD34+ cells derived from human umbilical cord blood before or after heat stroke has been shown to promote survival. We investigated whether CD34– human placenta-derived stem cells (PDMSCs) could improve survival following heat stroke in rats. Rats were subjected to heat stress (42°C for 98 min) to induce heat stroke. Intravenous administration of PDMSCs 1 day before or immediately after the onset of heat stroke improved survival by 60% and 20%, respectively. Pre-treatment with CD34− PDMSCs protected against heat stroke injury more effectively than that treatment after injury. PDMSCs treatment attenuated cerebrovascular dysfunction, the inflammatory response, and lipid peroxidation. These data suggest human PDMSCs protect against heat stroke injury in rats. Moreover, these effects do not require the presence of CD34+ cells.
Collapse
Affiliation(s)
- Willie Lin
- Meridigen Biotech Co., Ltd., Taipei, Taiwan
| | | | - Yu-Chin Su
- Meridigen Biotech Co., Ltd., Taipei, Taiwan
| | | | - Mao-Tsun Lin
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Zi-Hao Chen
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Ching-Ping Chang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan.,Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan.,The Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kao-Chang Lin
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan.,Department of Neurology, Chi Mei Medical Center, Tainan, Taiwan
| |
Collapse
|
26
|
Term vs. preterm cord blood cells for the prevention of preterm brain injury. Pediatr Res 2017; 82:1030-1038. [PMID: 28723885 DOI: 10.1038/pr.2017.170] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/15/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUNDWhite matter brain injury in preterm infants can induce neurodevelopmental deficits. Umbilical cord blood (UCB) cells demonstrate neuroprotective properties, but it is unknown whether cells obtained from preterm cord blood (PCB) vs. term cord blood (TCB) have similar efficacy. This study compared the ability of TCB vs. PCB cells to reduce white matter injury in preterm fetal sheep.METHODSHypoxia-ischemia (HI) was induced in fetal sheep (0.7 gestation) by 25 min umbilical cord occlusion. Allogeneic UCB cells from term or preterm sheep, or saline, were administered to the fetus at 12 h after HI. The fetal brain was collected at 10-day post HI for assessment of white matter neuropathology.RESULTSHI (n=7) induced cell death and microglial activation and reduced total oligodendrocytes and CNPase+myelin protein in the periventricular white matter and internal capsule when compared with control (n=10). Administration of TCB or PCB cells normalized white matter density and reduced cell death and microgliosis (P<0.05). PCB prevented upregulation of plasma tumor necrosis factor (TNF)-a, whereas TCB increased anti-inflammatory interleukin (IL)-10 (P<0.05). TCB, but not PCB, reduced circulating oxidative stress.CONCLUSIONSTCB and PCB cells reduced preterm HI-induced white matter injury, primarily via anti-inflammatory actions. The secondary mechanisms of neuroprotection appear different following TCB vs. PCB administration.
Collapse
|
27
|
Transplantation of Human Umbilical Cord Blood Mononuclear Cells Attenuated Ischemic Injury in MCAO Rats via Inhibition of NF-κB and NLRP3 Inflammasome. Neuroscience 2017; 369:314-324. [PMID: 29175152 DOI: 10.1016/j.neuroscience.2017.11.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 11/15/2017] [Accepted: 11/16/2017] [Indexed: 12/28/2022]
Abstract
Accumulated evidence displayed that transplantation of stem cells may be a promising approach for the treatment of neurological disorders. However, the underlying mechanisms remain to be well elucidated. Moreover, some investigators cannot reproduce similar results as the previous. The present results showed that transplantation of fresh human umbilical cord blood mononuclear cells (cbMNCs) attenuated ischemic damage in middle cerebral artery occlusion (MCAO) rats, accompanied with improvement of neurologic deficits, learning and memory function. The increase in neovascularization and related molecules such as vascular endothelial growth factor (VEGF), Angiopoietin-1 (Ang-1) and endothelium-specific receptor tyrosine kinase 2 (Tie-2) in the injured brain was observed in cbMNCs-treated rats. Moreover, nuclear factor-κB (NF-κB) activation and nucleotide binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome were also inhibited by the cells graft, resulting in reduction in cleaved caspase-1 and mature interleukin-1β (IL-1β) content. These results suggested that the protective actions of the cells on the cerebral ischemia may be related to inhibition of NF-κB pathway and NLRP3 inflammasome.
Collapse
|
28
|
Cui LL, Nitzsche F, Pryazhnikov E, Tibeykina M, Tolppanen L, Rytkönen J, Huhtala T, Mu JW, Khiroug L, Boltze J, Jolkkonen J. Integrin α4 Overexpression on Rat Mesenchymal Stem Cells Enhances Transmigration and Reduces Cerebral Embolism After Intracarotid Injection. Stroke 2017; 48:2895-2900. [PMID: 28916665 DOI: 10.1161/strokeaha.117.017809] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/03/2017] [Accepted: 08/15/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND PURPOSE Very late antigen-4 (integrin α4β1)/vascular cell adhesion molecule-1 mediates leukocyte trafficking and transendothelial migration after stroke. Mesenchymal stem cells (MSCs) typically express integrin β1 but insufficient ITGA4 (integrin α4), which limits their homing after intravascular transplantation. We tested whether ITGA4 overexpression on MSCs increases cerebral homing after intracarotid transplantation and reduces MSC-borne cerebral embolism. METHODS Rat MSCs were lentivirally transduced to overexpress ITGA4. In vitro transendothelial migration was assessed using a Boyden chamber assay. Male Wistar rats intracarotidly received 0.5×106 control or modified MSCs 24 hours after sham or stroke surgery. In vivo behavior of MSCs in the cerebral vasculature was observed by intravital microscopy and single-photon emission computed tomography for up to 72 hours. RESULTS Transendothelial migration of ITGA4-overexpressing MSCs was increased in vitro. MSCs were passively entrapped in microvessels in vivo and occasionally formed large cell aggregates causing local blood flow interruptions. MSCs were rarely found in perivascular niches or parenchyma at 72 hours post-transplantation, but ITGA4 overexpression significantly decreased cell aggregation and ameliorated the evoked cerebral embolism in stroke rats. CONCLUSIONS ITGA4 overexpression on MSCs enhances transendothelial migration in vitro, but not in vivo, although it improves safety after intracarotid transplantation into stroke rats.
Collapse
Affiliation(s)
- Li-Li Cui
- From the Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland (L.-l.C., J.-w.M., J.J.); Department of Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (F.N.); McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA (F.N.); In Vivo Brain Microscopy Unit, Neuroscience Center, University of Helsinki, Finland (E.P., M.T., L.K.); Charles River DRS Finland, Kuopio (L.T., J.R., T.H.); Department of Translational Medicine and Cell Technology, Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck, Germany (J.B.); and Neurocenter, Kuopio University Hospital, Finland (J.J.)
| | - Franziska Nitzsche
- From the Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland (L.-l.C., J.-w.M., J.J.); Department of Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (F.N.); McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA (F.N.); In Vivo Brain Microscopy Unit, Neuroscience Center, University of Helsinki, Finland (E.P., M.T., L.K.); Charles River DRS Finland, Kuopio (L.T., J.R., T.H.); Department of Translational Medicine and Cell Technology, Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck, Germany (J.B.); and Neurocenter, Kuopio University Hospital, Finland (J.J.)
| | - Evgeny Pryazhnikov
- From the Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland (L.-l.C., J.-w.M., J.J.); Department of Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (F.N.); McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA (F.N.); In Vivo Brain Microscopy Unit, Neuroscience Center, University of Helsinki, Finland (E.P., M.T., L.K.); Charles River DRS Finland, Kuopio (L.T., J.R., T.H.); Department of Translational Medicine and Cell Technology, Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck, Germany (J.B.); and Neurocenter, Kuopio University Hospital, Finland (J.J.)
| | - Marina Tibeykina
- From the Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland (L.-l.C., J.-w.M., J.J.); Department of Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (F.N.); McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA (F.N.); In Vivo Brain Microscopy Unit, Neuroscience Center, University of Helsinki, Finland (E.P., M.T., L.K.); Charles River DRS Finland, Kuopio (L.T., J.R., T.H.); Department of Translational Medicine and Cell Technology, Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck, Germany (J.B.); and Neurocenter, Kuopio University Hospital, Finland (J.J.)
| | - Laura Tolppanen
- From the Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland (L.-l.C., J.-w.M., J.J.); Department of Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (F.N.); McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA (F.N.); In Vivo Brain Microscopy Unit, Neuroscience Center, University of Helsinki, Finland (E.P., M.T., L.K.); Charles River DRS Finland, Kuopio (L.T., J.R., T.H.); Department of Translational Medicine and Cell Technology, Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck, Germany (J.B.); and Neurocenter, Kuopio University Hospital, Finland (J.J.)
| | - Jussi Rytkönen
- From the Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland (L.-l.C., J.-w.M., J.J.); Department of Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (F.N.); McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA (F.N.); In Vivo Brain Microscopy Unit, Neuroscience Center, University of Helsinki, Finland (E.P., M.T., L.K.); Charles River DRS Finland, Kuopio (L.T., J.R., T.H.); Department of Translational Medicine and Cell Technology, Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck, Germany (J.B.); and Neurocenter, Kuopio University Hospital, Finland (J.J.)
| | - Tuulia Huhtala
- From the Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland (L.-l.C., J.-w.M., J.J.); Department of Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (F.N.); McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA (F.N.); In Vivo Brain Microscopy Unit, Neuroscience Center, University of Helsinki, Finland (E.P., M.T., L.K.); Charles River DRS Finland, Kuopio (L.T., J.R., T.H.); Department of Translational Medicine and Cell Technology, Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck, Germany (J.B.); and Neurocenter, Kuopio University Hospital, Finland (J.J.)
| | - Jing-Wei Mu
- From the Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland (L.-l.C., J.-w.M., J.J.); Department of Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (F.N.); McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA (F.N.); In Vivo Brain Microscopy Unit, Neuroscience Center, University of Helsinki, Finland (E.P., M.T., L.K.); Charles River DRS Finland, Kuopio (L.T., J.R., T.H.); Department of Translational Medicine and Cell Technology, Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck, Germany (J.B.); and Neurocenter, Kuopio University Hospital, Finland (J.J.)
| | - Leonard Khiroug
- From the Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland (L.-l.C., J.-w.M., J.J.); Department of Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (F.N.); McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA (F.N.); In Vivo Brain Microscopy Unit, Neuroscience Center, University of Helsinki, Finland (E.P., M.T., L.K.); Charles River DRS Finland, Kuopio (L.T., J.R., T.H.); Department of Translational Medicine and Cell Technology, Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck, Germany (J.B.); and Neurocenter, Kuopio University Hospital, Finland (J.J.)
| | - Johannes Boltze
- From the Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland (L.-l.C., J.-w.M., J.J.); Department of Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (F.N.); McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA (F.N.); In Vivo Brain Microscopy Unit, Neuroscience Center, University of Helsinki, Finland (E.P., M.T., L.K.); Charles River DRS Finland, Kuopio (L.T., J.R., T.H.); Department of Translational Medicine and Cell Technology, Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck, Germany (J.B.); and Neurocenter, Kuopio University Hospital, Finland (J.J.)
| | - Jukka Jolkkonen
- From the Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland (L.-l.C., J.-w.M., J.J.); Department of Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (F.N.); McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA (F.N.); In Vivo Brain Microscopy Unit, Neuroscience Center, University of Helsinki, Finland (E.P., M.T., L.K.); Charles River DRS Finland, Kuopio (L.T., J.R., T.H.); Department of Translational Medicine and Cell Technology, Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck, Germany (J.B.); and Neurocenter, Kuopio University Hospital, Finland (J.J.).
| |
Collapse
|
29
|
Huang L, Liu Y, Lu J, Cerqueira B, Misra V, Duong TQ. Intraarterial transplantation of human umbilical cord blood mononuclear cells in hyperacute stroke improves vascular function. Stem Cell Res Ther 2017; 8:74. [PMID: 28330501 PMCID: PMC5361847 DOI: 10.1186/s13287-017-0529-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 01/18/2017] [Accepted: 03/04/2017] [Indexed: 12/20/2022] Open
Abstract
Background Human umbilical cord blood (hUCB) cell therapy is a promising treatment for ischemic stroke. The effects of hyperacute stem cell transplantation on cerebrovascular function in ischemic stroke are, however, not well understood. This study evaluated the effects of hyperacute intraarterial transplantation of hUCB mononuclear cells (MNCs) on cerebrovascular function in stroke rats using serial magnetic resonance imaging (MRI). Methods HUCB MNCs or vehicle were administered to stroke rats via the internal carotid artery immediately after reperfusion at 60 min following ischemia onset. Lesion volumes were longitudinally evaluated by MRI on days 0, 2, 14, and 28 after stroke, accompanied by behavioral tests. Cerebral blood flow (CBF) and cerebrovascular reactivity were measured by perfusion MRI and CO2 functional MRI (fMRI) at 28 days post-stroke; corresponding vascular morphological changes were also detected by immunohistology in the same animals. Results We found that CBF to the stroke-affected region at 28 days was improved (normalized CBF value: 1.41 ± 0.30 versus 0.49 ± 0.07) by intraarterial transplantation of hUCB MNCs in the hyperacute stroke phase, compared to vehicle control. Cerebrovascular reactivity within the stroke-affected area, measured by CBF fMRI, was also increased (35.2 ± 3.5% versus 12.8 ± 4.3%), as well as the corresponding cerebrovascular density. Some engrafted cells appeared with microvascular-like morphology and stained positive for von Willebrand Factor (an endothelial cell marker), suggesting they differentiated into endothelial cells. Some engrafted cells also connected to host endothelial cells, suggesting they interacted with the host vasculature. Compared to the vehicle group, infarct volume at 28 days in the stem cell treated group was significantly smaller (160.9 ± 15.7 versus 231.2 ± 16.0 mm3); behavioral deficits were also markedly reduced by stem cell treatment at day 28 (19.5 ± 1.0% versus 30.7 ± 4.7% on the foot fault test; 68.2 ± 4.6% versus 86.6 ± 5.8% on the cylinder test). More tissue within initial perfusion-diffusion mismatch was rescued in the treatment group. Conclusions Intraarterial hUCB MNC transplantation during the hyperacute phase of ischemic stroke improved cerebrovascular function and reduced behavioral deficits and infarct volume.
Collapse
Affiliation(s)
- Lei Huang
- Research Imaging Institute, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Yichu Liu
- Department of Biomedical Engineering, University of Texas, San Antonio, Texas, USA
| | - Jianfei Lu
- Research Imaging Institute, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Bianca Cerqueira
- Research Imaging Institute, University of Texas Health Science Center, San Antonio, Texas, USA.,Department of Biomedical Engineering, University of Texas, San Antonio, Texas, USA
| | - Vivek Misra
- Department of Neurology, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Timothy Q Duong
- Research Imaging Institute, University of Texas Health Science Center, San Antonio, Texas, USA. .,Radiology, Stony Brook Medicine, Stony Brook, NY, USA.
| |
Collapse
|
30
|
Sun GD, Chen Y, Zhou ZG, Yang SX, Zhong C, Li ZZ. A progressive compression model of thoracic spinal cord injury in mice: function assessment and pathological changes in spinal cord. Neural Regen Res 2017; 12:1365-1374. [PMID: 28966654 PMCID: PMC5607834 DOI: 10.4103/1673-5374.213693] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Non-traumatic injury accounts for approximately half of clinical spinal cord injury, including chronic spinal cord compression. However, previous rodent spinal cord compression models are mainly designed for rats, few are available for mice. Our aim is to develop a thoracic progressive compression mice model of spinal cord injury. In this study, adult wild-type C57BL/6 mice were divided into two groups: in the surgery group, a screw was inserted at T9 lamina to compress the spinal cord, and the compression was increased by turning it further into the canal (0.2 mm) post-surgery every 2 weeks up to 8 weeks. In the control group, a hole was drilled into the lamina without inserting a screw. The results showed that Basso Mouse Scale scores were lower and gait worsened. In addition, the degree of hindlimb dysfunction in mice was consistent with the degree of spinal cord compression. The number of motor neurons in the anterior horn of the spinal cord was reduced in all groups of mice, whereas astrocytes and microglia were gradually activated and proliferated. In conclusion, this progressive compression of thoracic spinal cord injury in mice is a preferable model for chronic progressive spinal cord compression injury.
Collapse
Affiliation(s)
- Guo-Dong Sun
- Department of Orthopedics, First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Yan Chen
- Department of Orthopedics, First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Zhi-Gang Zhou
- Department of Orthopedics, First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Shu-Xian Yang
- Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, Guangdong Province, China
| | - Cheng Zhong
- Department of Traumatology and Plastic Surgery, The Affiliated Jiangmen Traditional Chinese Medicine Hospital of Jinan University, Jiangmen, Guangdong Province, China
| | - Zhi-Zhong Li
- Department of Orthopedics, First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China.,Department of Orthopedics, Heyuan People's Hospital (Heyuan Affiliated Hospital of Jinan University), Heyuan, Guangdong Province, China
| |
Collapse
|
31
|
Microvesicles from brain-extract-treated mesenchymal stem cells improve neurological functions in a rat model of ischemic stroke. Sci Rep 2016; 6:33038. [PMID: 27609711 PMCID: PMC5016792 DOI: 10.1038/srep33038] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 08/17/2016] [Indexed: 12/16/2022] Open
Abstract
Transplantation of mesenchymal stem cells (MSCs) was reported to improve functional outcomes in a rat model of ischemic stroke, and subsequent studies suggest that MSC-derived microvesicles (MVs) can replace the beneficial effects of MSCs. Here, we evaluated three different MSC-derived MVs, including MVs from untreated MSCs (MSC-MVs), MVs from MSCs treated with normal rat brain extract (NBE-MSC-MVs), and MVs from MSCs treated with stroke-injured rat brain extract (SBE-MSC-MVs), and tested their effects on ischemic brain injury induced by permanent middle cerebral artery occlusion (pMCAO) in rats. NBE-MSC-MVs and SBE-MSC-MVs had significantly greater efficacy than MSC-MVs for ameliorating ischemic brain injury with improved functional recovery. We found similar profiles of key signalling proteins in NBE-MSC-MVs and SBE-MSC-MVs, which account for their similar therapeutic efficacies. Immunohistochemical analyses suggest that brain-extract—treated MSC-MVs reduce inflammation, enhance angiogenesis, and increase endogenous neurogenesis in the rat brain. We performed mass spectrometry proteomic analyses and found that the total proteomes of brain-extract—treated MSC-MVs are highly enriched for known vesicular proteins. Notably, MSC-MV proteins upregulated by brain extracts tend to be modular for tissue repair pathways. We suggest that MSC-MV proteins stimulated by the brain microenvironment are paracrine effectors that enhance MSC therapy for stroke injury.
Collapse
|
32
|
Preterm white matter brain injury is prevented by early administration of umbilical cord blood cells. Exp Neurol 2016; 283:179-87. [PMID: 27317990 DOI: 10.1016/j.expneurol.2016.06.017] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 06/13/2016] [Accepted: 06/14/2016] [Indexed: 12/29/2022]
Abstract
Infants born very preterm are at high risk for neurological deficits including cerebral palsy. In this study we assessed the neuroprotective effects of umbilical cord blood cells (UCBCs) and optimal administration timing in a fetal sheep model of preterm brain injury. 50 million allogeneic UCBCs were intravenously administered to fetal sheep (0.7 gestation) at 12h or 5d after acute hypoxia-ischemia (HI) induced by umbilical cord occlusion. The fetal brains were collected at 10d after HI. HI (n=7) was associated with reduced number of oligodendrocytes (Olig2+) and myelin density (CNPase+), and increased density of activated microglia (Iba-1+) in cerebral white matter compared to control fetuses (P<0.05). UCBCs administered at 12h, but not 5d after HI, significantly protected white matter structures and suppressed cerebral inflammation. Activated microglial density showed a correlation with decreasing oligodendrocyte number (P<0.001). HI caused cell death (TUNEL+) in the internal capsule and cell proliferation (Ki-67+) in the subventricular zone compared to control (P<0.05), while UCBCs at 12h or 5d ameliorated these effects. Additionally, UCBCs at 12h induced a significant systemic increase in interleukin-10 at 10d, and reduced oxidative stress (malondialdehyde) following HI (P<0.05). UCBC administration at 12h after HI reduces preterm white matter injury, via anti-inflammatory and antioxidant actions.
Collapse
|
33
|
Xiong XY, Liu L, Yang QW. Functions and mechanisms of microglia/macrophages in neuroinflammation and neurogenesis after stroke. Prog Neurobiol 2016; 142:23-44. [PMID: 27166859 DOI: 10.1016/j.pneurobio.2016.05.001] [Citation(s) in RCA: 464] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 04/20/2016] [Accepted: 05/01/2016] [Indexed: 02/08/2023]
Abstract
Microglia/macrophages are the major immune cells involved in the defence against brain damage. Their morphology and functional changes are correlated with the release of danger signals induced by stroke. These cells are normally responsible for clearing away dead neural cells and restoring neuronal functions. However, when excessively activated by the damage-associated molecular patterns following stroke, they can produce a large number of proinflammatory cytokines that can disrupt neural cells and the blood-brain barrier and influence neurogenesis. These effects indicate the important roles of microglia/macrophages in the pathophysiological processes of stroke. However, the modifiable and adaptable nature of microglia/macrophages may also be beneficial for brain repair and not just result in damage. These distinct roles may be attributed to the different microglia/macrophage phenotypes because the M1 population is mainly destructive, while the M2 population is neuroprotective. Additionally, different gene expression signature changes in microglia/macrophages have been found in diverse inflammatory milieus. These biofunctional features enable dual roles for microglia/macrophages in brain damage and repair. Currently, it is thought that the proper inflammatory milieu may provide a suitable microenvironment for neurogenesis; however, detailed mechanisms underlying the inflammatory responses that initiate or inhibit neurogenesis remain unknown. This review summarizes recent progress concerning the mechanisms involved in brain damage, repair and regeneration related to microglia/macrophage activation and phenotype transition after stroke. We also argue that future translational studies should be targeting multiple key regulating molecules to improve brain repair, which should be accompanied by the concept of a "therapeutic time window" for sequential therapies.
Collapse
Affiliation(s)
- Xiao-Yi Xiong
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Xinqiao zhengjie No.183, Shapingba District Chongqing, 400037, China
| | - Liang Liu
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Xinqiao zhengjie No.183, Shapingba District Chongqing, 400037, China
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Xinqiao zhengjie No.183, Shapingba District Chongqing, 400037, China.
| |
Collapse
|
34
|
Hocum Stone LL, Xiao F, Rotschafer J, Nan Z, Juliano M, Sanberg CD, Sanberg PR, Kuzmin-Nichols N, Grande A, Cheeran MCJ, Low WC. Amelioration of Ischemic Brain Injury in Rats With Human Umbilical Cord Blood Stem Cells: Mechanisms of Action. Cell Transplant 2016; 25:1473-88. [PMID: 26996530 DOI: 10.3727/096368916x691277] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Despite the high prevalence and devastating outcome, there remain a few options for treatment of ischemic stroke. Currently available treatments are limited by a short time window for treatment and marginal efficacy when used. We have tested a human umbilical cord blood-derived stem cell line that has been shown to result in a significant reduction in stroke infarct volume as well as improved functional recovery following stroke in the rat. In the present study we address the mechanism of action and compared the therapeutic efficacy of high- versus low-passage nonhematopoietic umbilical cord blood stem cells (nh-UCBSCs). Using the middle cerebral arterial occlusion (MCAo) model of stroke in Sprague-Dawley rats, we administered nh-UCBSC by intravenous (IV) injection 2 days following stroke induction. These human cells were injected into rats without any immune suppression, and no adverse reactions were detected. Both behavioral and histological analyses have shown that the administration of these cells reduces the infarct volume by 50% as well as improves the functional outcome of these rats following stroke for both high- and low-passaged nh-UCBSCs. Flow cytometry analysis of immune cells present in the brains of normal rats, rats with ischemic brain injury, and ischemic animals with nh-UCBSC treatment confirmed infiltration of macrophages and T cells consequent to ischemia and reduction to normal levels with nh-UCBSC treatment. Flow cytometry also revealed a restoration of normal levels of microglia in the brain following treatment. These data suggest that nh-UCBSCs may act by inhibiting immune cell migration into the brain from the periphery and possibly by inhibition of immune cell activation within the brain. nh-UCBSCs exhibit great potential for treatment of stroke, including the fact that they are associated with an increased therapeutic time window, no known ill-effects, and that they can be expanded to high numbers for, and stored for, treatment.
Collapse
|
35
|
Savitz SI, Parsha K. Enhancing Stroke Recovery with Cellular Therapies. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00060-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
36
|
Jablonska A, Drela K, Wojcik-Stanaszek L, Janowski M, Zalewska T, Lukomska B. Short-Lived Human Umbilical Cord-Blood-Derived Neural Stem Cells Influence the Endogenous Secretome and Increase the Number of Endogenous Neural Progenitors in a Rat Model of Lacunar Stroke. Mol Neurobiol 2015; 53:6413-6425. [PMID: 26607630 PMCID: PMC5085993 DOI: 10.1007/s12035-015-9530-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 11/08/2015] [Indexed: 12/12/2022]
Abstract
Stroke is the leading cause of severe disability, and lacunar stroke is related to cognitive decline and hemiparesis. There is no effective treatment for the majority of patients with stroke. Thus, stem cell-based regenerative medicine has drawn a growing body of attention due to the capabilities for trophic factor expression and neurogenesis enhancement. Moreover, it was shown in an experimental autoimmune encephalomyelitis (EAE) model that even short-lived stem cells can be therapeutic, and we have previously observed that phenomenon indirectly. Here, in a rat model of lacunar stroke, we investigated the molecular mechanisms underlying the positive therapeutic effects of short-lived human umbilical cord-blood-derived neural stem cells (HUCB-NSCs) through the distinct measurement of exogenous human and endogenous rat trophic factors. We have also evaluated neurogenesis and metalloproteinase activity as cellular components of therapeutic activity. As expected, we observed an increased proliferation and migration of progenitors, as well as metalloproteinase activity up to 14 days post transplantation. These changes were most prominent at the 7-day time point when we observed 30 % increases in the number of bromodeoxyuridine (BrdU)-positive cells in HUCB-NSC transplanted animals. The expression of human trophic factors was present until 7 days post transplantation, which correlated well with the survival of the human graft. For these 7 days, the level of messenger RNA (mRNA) in the analyzed trophic factors was from 300-fold for CNTF to 10,000-fold for IGF, much higher compared to constitutive expression in HUCB-NSCs in vitro. What is interesting is that there was no increase in the expression of rat trophic factors during the human graft survival, compared to that in non-transplanted animals. However, there was a prolongation of a period of increased trophic expression until 14 days post transplantation, while, in non-transplanted animals, there was a significant drop in rat trophic expression at that time point. We conclude that the positive therapeutic effect of short-lived stem cells may be related to the net increase in the amount of trophic factors (rat + human) until graft death and to the prolonged increase in rat trophic factor expression subsequently.
Collapse
Affiliation(s)
- Anna Jablonska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Drela
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Luiza Wojcik-Stanaszek
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Miroslaw Janowski
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.,Cellular Imaging Section, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
37
|
Kadam SD, Chen H, Markowitz GJ, Raja S, George S, Verina T, Shotwell E, Loechelt B, Johnston MV, Kamani N, Fatemi A, Comi AM. Systemic injection of CD34(+)-enriched human cord blood cells modulates poststroke neural and glial response in a sex-dependent manner in CD1 mice. Stem Cells Dev 2015; 24:51-66. [PMID: 25121827 DOI: 10.1089/scd.2014.0135] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Stroke in the developing brain is an important cause of neurological morbidity. We determined the impact of human cord blood-derived CD34(+)-enriched mononuclear cells (CBSC) intraperitoneally injected 48 h after an ischemic stroke at postnatal day 12 by evaluating poststroke neurogenic niche proliferation, glial response, and recovery in CD1 mice. Percent brain atrophy was quantified from Nissl-stained sections. Density of BrdU, Iba-1, and GFAP staining were quantified in the dentate gyrus and the subventricular zone (SVZ). Immunohistochemistry for human nuclear antibody, human mitochondrial antibody, and human CD34(+) cells was done on injured and uninjured brains from CBSC- and vehicle-treated mice. Developmental neurobehavioral milestones were evaluated pre- and post-treatment. No significant differences in stroke severity were noted between CBSC and vehicle-treated injured animals. With a 1×10(5) CBSC dose, there was a significant increase in subgranular zone (SGZ) proliferation in the CBSC-versus vehicle-treated stroke-injured male mice. SVZ glial fibrillary acidic protein (GFAP) expression was increased contralaterally in injured females treated with CBSC but suppressed in injured males. Significant negative correlations between severity of the stroke-injury and spleen weights, and between spleen weights and SGZ proliferation, and a positive correlation between GFAP expression and severity of brain injury were noted in the vehicle-treated injured mice but not in the CBSC-treated mice. GFAP expression and SVZ proliferation were positively correlated. In conclusion, neurogenic niche proliferation and glial brain responses to CBSC after neonatal stroke may involve interactions with the spleen and are sex dependent.
Collapse
Affiliation(s)
- Shilpa D Kadam
- 1 Department of Neurology and Developmental Medicine, Kennedy Krieger Research Institute , Baltimore, Maryland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Lee SH, Jin KS, Bang OY, Kim BJ, Park SJ, Lee NH, Yoo KH, Koo HH, Sung KW. Differential Migration of Mesenchymal Stem Cells to Ischemic Regions after Middle Cerebral Artery Occlusion in Rats. PLoS One 2015; 10:e0134920. [PMID: 26241653 PMCID: PMC4524688 DOI: 10.1371/journal.pone.0134920] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 07/15/2015] [Indexed: 12/16/2022] Open
Abstract
To evaluate the optimal timing of mesenchymal stem cell (MSC) transplantation following stroke, rats were transplanted with MSCs at 1 (D1), 4 (D4), and 7 days (D7) after middle cerebral artery occlusion (MCAo). Rats in the D1 group showed a better functional recovery than those in the D4 or D7 groups after MCAo. MSCs preferentially migrated to the cortex in the D1 group, while the MSCs in the D4 or D7 groups preferentially migrated to the striatum. Interestingly, the level of monocyte chemotactic protein-1 (MCP-1) in the cortex was highest at 1 day after MCAo, while the level of stromal cell-derived factor-1 (SDF-1) in the striatum was lowest at 1 day after MCAo and then increased over time. The pattern of MCP-1 and SDF-1 level changes according to the time after MCAo was consistent with in vivo and in vitro migration patterns of MSCs. The results suggest that an earlier MSC transplantation is associated with a better functional recovery after stroke, which could be explained by the preferential migration of MSCs to the cortex in the early transplantation group. The time-dependent differential expression of MCP-1 and SDF-1 between ischemic regions seemed to mediate the differential migration of MSCs. Highest level of MCP-1 at one day of stroke may induce preferential migration of MSCs to the cortex, then better functional improvement.
Collapse
Affiliation(s)
- Soo Hyun Lee
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyung Sil Jin
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Oh Young Bang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Byoung Joon Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Soo Jin Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Na Hee Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
39
|
Boltze J, Arnold A, Walczak P, Jolkkonen J, Cui L, Wagner DC. The Dark Side of the Force - Constraints and Complications of Cell Therapies for Stroke. Front Neurol 2015; 6:155. [PMID: 26257702 PMCID: PMC4507146 DOI: 10.3389/fneur.2015.00155] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/23/2015] [Indexed: 12/16/2022] Open
Abstract
Cell therapies are increasingly recognized as a promising option to augment the limited therapeutic arsenal available to fight ischemic stroke. During the last two decades, cumulating preclinical evidence has indicated a substantial efficacy for most cell treatment paradigms and first clinical trials are currently underway to assess safety and feasibility in patients. However, the strong and still unmet demand for novel stroke treatment options and exciting findings reported from experimental studies may have drawn our attention away from potential side effects related to cell therapies and the ways by which they are commonly applied. This review summarizes common and less frequent adverse events that have been discovered in preclinical and clinical investigations assessing cell therapies for stroke. Such adverse events range from immunological and neoplastic complications over seizures to cell clotting and cell-induced embolism. It also describes potential complications of clinically applicable administration procedures, detrimental interactions between therapeutic cells, and the pathophysiological environment that they are placed into, as well as problems related to cell manufacturing. Virtually each therapeutic intervention comes at a certain risk for complications. Side effects do therefore not generally compromise the value of cell treatments for stroke, but underestimating such complications might severely limit therapeutic safety and efficacy of cell treatment protocols currently under development. On the other hand, a better understanding will provide opportunities to further improve existing therapeutic strategies and might help to define those circumstances, under which an optimal effect can be realized. Hence, the review eventually discusses strategies and recommendations allowing us to prevent or at least balance potential complications in order to ensure the maximum therapeutic benefit at minimum risk for stroke patients.
Collapse
Affiliation(s)
- Johannes Boltze
- Department of Cell Therapy, Fraunhofer-Institute for Cell Therapy and Immunology , Leipzig , Germany ; Translational Center for Regenerative Medicine, University of Leipzig , Leipzig , Germany
| | - Antje Arnold
- Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine , Baltimore, MD , USA ; Institute for Cell Engineering, Johns Hopkins University , Baltimore, MD , USA
| | - Piotr Walczak
- Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine , Baltimore, MD , USA ; Institute for Cell Engineering, Johns Hopkins University , Baltimore, MD , USA
| | - Jukka Jolkkonen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland , Kuopio , Finland
| | - Lili Cui
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland , Kuopio , Finland
| | - Daniel-Christoph Wagner
- Department of Cell Therapy, Fraunhofer-Institute for Cell Therapy and Immunology , Leipzig , Germany
| |
Collapse
|
40
|
Gavins FNE, Smith HK. Cell tracking technologies for acute ischemic brain injury. J Cereb Blood Flow Metab 2015; 35:1090-9. [PMID: 25966948 PMCID: PMC4640284 DOI: 10.1038/jcbfm.2015.93] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 04/07/2015] [Accepted: 04/09/2015] [Indexed: 02/06/2023]
Abstract
Stem cell therapy has showed considerable potential in the treatment of stroke over the last decade. In order that these therapies may be optimized, the relative benefits of growth factor release, immunomodulation, and direct tissue replacement by therapeutic stem cells are widely under investigation. Fundamental to the progress of this research are effective imaging techniques that enable cell tracking in vivo. Direct analysis of the benefit of cell therapy includes the study of cell migration, localization, division and/or differentiation, and survival. This review explores the various imaging tools currently used in clinics and laboratories, addressing image resolution, long-term cell monitoring, imaging agents/isotopes, as well as safety and costs associated with each technique. Finally, burgeoning tracking techniques are discussed, with emphasis on multimodal imaging.
Collapse
Affiliation(s)
- Felicity NE Gavins
- Molecular and Cellular Physiology Department, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA
| | - Helen K Smith
- Molecular and Cellular Physiology Department, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA
| |
Collapse
|
41
|
Janowski M, Wagner DC, Boltze J. Stem Cell-Based Tissue Replacement After Stroke: Factual Necessity or Notorious Fiction? Stroke 2015; 46:2354-63. [PMID: 26106118 DOI: 10.1161/strokeaha.114.007803] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 04/28/2015] [Indexed: 01/09/2023]
Affiliation(s)
- Miroslaw Janowski
- From the Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research (M.J.) and Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering (M.J.), The Johns Hopkins University School of Medicine, Baltimore, MD; NeuroRepair Department (M.J.) and Department of Neurosurgery (M.J.), Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland; Department of Cell Therapy, Fraunhofer-Institute for Cell Therapy and Immunology, Translational Centre for Regenerative Medicine, Leipzig, Germany (D.-C.W., J.B.); and Stroke and Neurovascular Regulation Laboratory, Neuroscience Center at Massachussets General Hospital, Harvard Medical School, Stroke and Neurovascular Regulation Laboratory, Charlestown, MA (J.B.)
| | - Daniel-Christoph Wagner
- From the Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research (M.J.) and Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering (M.J.), The Johns Hopkins University School of Medicine, Baltimore, MD; NeuroRepair Department (M.J.) and Department of Neurosurgery (M.J.), Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland; Department of Cell Therapy, Fraunhofer-Institute for Cell Therapy and Immunology, Translational Centre for Regenerative Medicine, Leipzig, Germany (D.-C.W., J.B.); and Stroke and Neurovascular Regulation Laboratory, Neuroscience Center at Massachussets General Hospital, Harvard Medical School, Stroke and Neurovascular Regulation Laboratory, Charlestown, MA (J.B.)
| | - Johannes Boltze
- From the Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research (M.J.) and Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering (M.J.), The Johns Hopkins University School of Medicine, Baltimore, MD; NeuroRepair Department (M.J.) and Department of Neurosurgery (M.J.), Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland; Department of Cell Therapy, Fraunhofer-Institute for Cell Therapy and Immunology, Translational Centre for Regenerative Medicine, Leipzig, Germany (D.-C.W., J.B.); and Stroke and Neurovascular Regulation Laboratory, Neuroscience Center at Massachussets General Hospital, Harvard Medical School, Stroke and Neurovascular Regulation Laboratory, Charlestown, MA (J.B.).
| |
Collapse
|
42
|
Detante O, Jaillard A, Moisan A, Barbieux M, Favre I, Garambois K, Hommel M, Remy C. Biotherapies in stroke. Rev Neurol (Paris) 2014; 170:779-98. [DOI: 10.1016/j.neurol.2014.10.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 09/29/2014] [Accepted: 10/08/2014] [Indexed: 12/31/2022]
|
43
|
Lu H, Hu H, He Z, Han X, Chen J, Tu R. Therapeutic imaging window of cerebral infarction revealed by multisequence magnetic resonance imaging: An animal and clinical study. Neural Regen Res 2014; 7:2446-55. [PMID: 25337095 PMCID: PMC4200719 DOI: 10.3969/j.issn.1673-5374.2012.31.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 10/09/2012] [Indexed: 12/17/2022] Open
Abstract
In this study, we established a Wistar rat model of right middle cerebral artery occlusion and observed pathological imaging changes (T2-weighted imaging [T2WI], T2FLAIR, and diffusion-weighted imaging [DWI]) following cerebral infarction. The pathological changes were divided into three phases: early cerebral infarction, middle cerebral infarction, and late cerebral infarction. In the early cerebral infarction phase (less than 2 hours post-infarction), there was evidence of intracellular edema, which improved after reperfusion. This improvement was defined as the ischemic penumbra. In this phase, a high DWI signal and a low apparent diffusion coefficient were observed in the right basal ganglia region. By contrast, there were no abnormal T2WI and T2FLAIR signals. For the middle cerebral infarction phase (2–4 hours post-infarction), a mixed edema was observed. After reperfusion, there was a mild improvement in cell edema, while the angioedema became more serious. A high DWI signal and a low apparent diffusion coefficient signal were observed, and some rats showed high T2WI and T2FLAIR signals. For the late cerebral infarction phase (4–6 hours post-infarction), significant angioedema was visible in the infarction site. After reperfusion, there was a significant increase in angioedema, while there was evidence of hemorrhage and necrosis. A mixed signal was observed on DWI, while a high apparent diffusion coefficient signal, a high T2WI signal, and a high T2FLAIR signal were also observed. All 86 cerebral infarction patients were subjected to T2WI, T2FLAIR, and DWI. MRI results of clinic data similar to the early infarction phase of animal experiments were found in 51 patients, for which 10 patients (10/51) had an onset time greater than 6 hours. A total of 35 patients had MRI results similar to the middle and late infarction phase of animal experiments, of which eight patients (8/35) had an onset time less than 6 hours. These data suggest that defining the “therapeutic time window” as the time 6 hours after infarction may not be suitable for all patients. Integrated application of MRI sequences including T2WI, T2FLAIR, DW-MRI, and apparent diffusion coefficient mapping should be used to examine the ischemic penumbra, which may provide valuable information for identifying the “therapeutic time window”.
Collapse
Affiliation(s)
- Hong Lu
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University (Haikou Municipal People's Hospital), Haikou 570208, Hainan Province, China
| | - Hui Hu
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University (Haikou Municipal People's Hospital), Haikou 570208, Hainan Province, China
| | - Zhanping He
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University (Haikou Municipal People's Hospital), Haikou 570208, Hainan Province, China
| | - Xiangjun Han
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University (Haikou Municipal People's Hospital), Haikou 570208, Hainan Province, China
| | - Jing Chen
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University (Haikou Municipal People's Hospital), Haikou 570208, Hainan Province, China
| | - Rong Tu
- Department of Radiology, Affiliated Hospital of Hainan Medical University, Haikou 570102, Hainan Province, China
| |
Collapse
|
44
|
Pereira-Cunha FG, Duarte ASS, Reis-Alves SC, Olalla Saad ST, Metze K, Lorand-Metze I, Luzo ÂCM. Umbilical cord blood CD34(+) stem cells and other mononuclear cell subtypes processed up to 96 h from collection and stored at room temperature maintain a satisfactory functionality for cell therapy. Vox Sang 2014; 108:72-81. [PMID: 25333825 DOI: 10.1111/vox.12199] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 06/13/2014] [Accepted: 08/05/2014] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND OBJECTIVES Umbilical cord blood (UCB) is a good stem cell source for cell therapy. We recently demonstrated that cord blood mononuclear cell (MNCs) subtypes were viable and functional until 96 h after collection, even stored at room temperature. Now, we analyzed the viability and functionality of the cells before and after cryopreservation. MATERIALS AND METHODS Twenty UCB units were analyzed at 24 and 96 h after collection, frozen for 6 months, thawed and re-evaluated. MNCs were analyzed by flow cytometry, viability by 7-AAD and clonogenic assays (CFU) were performed. RESULTS After 96 h of storage, no substantial loss of MNC was found (median 7.320 × 10(6 ) × 6.05 × 10(6) ). Percentage and viability CD34(+) cells, B-cell precursors and mesenchymal stem cells were not affected. However, mature B and T lymphocytes as well as granulocytes had a substantial loss. CFU growth was observed in all samples. Prefreezing storage of 96 h was associated with a relative loss of colony formation (median 12%). Postthaw, this loss had a median of 49% (24 h samples) to 56% (96 h samples). CONCLUSION The delay of 96 h before UCB processing is possible, without a prohibitive impairment of CD34(+) loss in number and functionality.
Collapse
Affiliation(s)
- F G Pereira-Cunha
- Flow Cytometry Laboratory, Haematology Hemotherapy Center, University of Campinas, Campinas, Brazil
| | | | | | | | | | | | | |
Collapse
|
45
|
Skardelly M, Gaber K, Burdack S, Scheidt F, Schuhmann MU, Hilbig H, Meixensberger J, Boltze J. Transient but not permanent benefit of neuronal progenitor cell therapy after traumatic brain injury: potential causes and translational consequences. Front Cell Neurosci 2014; 8:318. [PMID: 25352780 PMCID: PMC4196631 DOI: 10.3389/fncel.2014.00318] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 09/23/2014] [Indexed: 01/16/2023] Open
Abstract
Background: Numerous studies have reported a beneficial impact of neural progenitor cell transplantation on functional outcome after traumatic brain injury (TBI) during short and medium follow-up periods. However, our knowledge regarding long-term functional effects is fragmentary while a direct comparison between local and systemic transplantation is missing so far. Objectives: This study investigated the long-term (12 week) impact of human fetal neuronal progenitor cell (hNPC) transplantation 24 h after severe TBI in rats. Methods: Cells were either transplanted stereotactically (1 × 105) into the putamen or systemically (5 × 105) via the tail vein. Control animals received intravenous transplantation of vehicle solution. Results: An overall functional benefit was observed after systemic, but not local hNPC transplantation by area under the curve analysis (p < 0.01). Surprisingly, this effect vanished during later stages after TBI with all groups exhibiting comparable functional outcomes 84 days after TBI. Investigation of cell-mediated inflammatory processes revealed increasing microglial activation and macrophage presence during these stages, which was statistically significant after systemic cell administration (p < 0.05). Intracerebral hNPC transplantation slightly diminished astrogliosis in perilesional areas (p < 0.01), but did not translate into a permanent functional benefit. No significant effects on angiogenesis were observed among the groups. Conclusion: Our results suggest the careful long-term assessment of cell therapies for TBI, as well as to identify potential long-term detrimental effects of such therapies before moving on to clinical trials. Moreover, immunosuppressive protocols, though widely used, should be rigorously assessed for their applicability in the respective setup.
Collapse
Affiliation(s)
- Marco Skardelly
- Department of Neurosurgery, University of Leipzig Leipzig, Germany
| | - Khaled Gaber
- Department of Neurosurgery, University of Leipzig Leipzig, Germany
| | - Swen Burdack
- Department of Neurosurgery, University of Leipzig Leipzig, Germany
| | | | | | | | | | - Johannes Boltze
- Fraunhofer Institute for Cell Therapy and Immunology and Translational Centre for Regenerative Medicine, University of Leipzig Leipzig, Germany ; Stroke and Neurovascular Regulation Laboratory, Massachussets General Hospital and Harvard Medical School Charlestown, MA, USA
| |
Collapse
|
46
|
Li J, McDonald CA, Fahey MC, Jenkin G, Miller SL. Could cord blood cell therapy reduce preterm brain injury? Front Neurol 2014; 5:200. [PMID: 25346720 PMCID: PMC4191167 DOI: 10.3389/fneur.2014.00200] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 09/19/2014] [Indexed: 12/25/2022] Open
Abstract
Major advances in neonatal care have led to significant improvements in survival rates for preterm infants, but this occurs at a cost, with a strong causal link between preterm birth and neurological deficits, including cerebral palsy (CP). Indeed, in high-income countries, up to 50% of children with CP were born preterm. The pathways that link preterm birth and brain injury are complex and multifactorial, but it is clear that preterm birth is strongly associated with damage to the white matter of the developing brain. Nearly 90% of preterm infants who later develop spastic CP have evidence of periventricular white matter injury. There are currently no treatments targeted at protecting the immature preterm brain. Umbilical cord blood (UCB) contains a diverse mix of stem and progenitor cells, and is a particularly promising source of cells for clinical applications, due to ethical and practical advantages over other potential therapeutic cell types. Recent studies have documented the potential benefits of UCB cells in reducing brain injury, particularly in rodent models of term neonatal hypoxia–ischemia. These studies indicate that UCB cells act via anti-inflammatory and immuno-modulatory effects, and release neurotrophic growth factors to support the damaged and surrounding brain tissue. The etiology of brain injury in preterm-born infants is less well understood than in term infants, but likely results from episodes of hypoperfusion, hypoxia–ischemia, and/or inflammation over a developmental period of white matter vulnerability. This review will explore current knowledge about the neuroprotective actions of UCB cells and their potential to ameliorate preterm brain injury through neonatal cell administration. We will also discuss the characteristics of UCB-derived from preterm and term infants for use in clinical applications.
Collapse
Affiliation(s)
- Jingang Li
- The Ritchie Centre, MIMR-PHI Institute , Clayton, VIC , Australia
| | | | - Michael C Fahey
- The Ritchie Centre, MIMR-PHI Institute , Clayton, VIC , Australia ; Department of Paediatrics, Monash University , Clayton, VIC , Australia
| | - Graham Jenkin
- The Ritchie Centre, MIMR-PHI Institute , Clayton, VIC , Australia ; Department of Obstetrics and Gynaecology, Monash University , Clayton, VIC , Australia
| | - Suzanne L Miller
- The Ritchie Centre, MIMR-PHI Institute , Clayton, VIC , Australia ; Department of Obstetrics and Gynaecology, Monash University , Clayton, VIC , Australia
| |
Collapse
|
47
|
Monocytes are essential for the neuroprotective effect of human cord blood cells following middle cerebral artery occlusion in rat. Mol Cell Neurosci 2014; 59:76-84. [PMID: 24472845 DOI: 10.1016/j.mcn.2014.01.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 01/17/2014] [Accepted: 01/20/2014] [Indexed: 12/17/2022] Open
Abstract
Systemic administration of human umbilical cord blood (HUCB) mononuclear cells (MNC) following middle cerebral artery occlusion (MCAO) in the rat reduces infarct size and, more importantly, restores motor function. The HUCB cell preparation is composed of immature T-cells, B-cells, monocytes and stem cells. In this study we examined whether the beneficial effects of HUCB injection were attributable to one of these cell types. Male Sprague Dawley rats underwent permanent MCAO followed 48 h later by intravenous administration of HUCB MNC preparations depleted of either CD14(+) monocytes, CD133(+) stem cells, CD2(+) T-cells or CD19(+) B cells. Motor function was measured prior to MCAO and 30 days post-stroke. When CD14(+) monocytes were depleted from the HUCB MNC, activity and motor asymmetry were similar to the MCAO only treated animals. Monocyte depletion prevented HUCB cell treatment from reducing infarct size while monocyte enrichment was sufficient to reduce infarct size. Administration of monocyte-depleted HUCB cells did not suppress Iba1 labeling of microglia in the infarcted area relative to treatment with the whole HUCB preparation. These data demonstrate that the HUCB monocytes provide the majority of the efficacy in reducing infarct volume and promoting functional recovery.
Collapse
|
48
|
Transplantation of cryopreserved human umbilical cord blood mononuclear cells does not induce sustained recovery after experimental stroke in spontaneously hypertensive rats. J Cereb Blood Flow Metab 2014; 34:e1-9. [PMID: 24169850 PMCID: PMC3887359 DOI: 10.1038/jcbfm.2013.185] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 09/25/2013] [Accepted: 09/26/2013] [Indexed: 01/13/2023]
Abstract
Previous studies have highlighted the enormous potential of cell-based therapies for stroke not only to prevent ischemic brain damage, but also to amplify endogenous repair processes. Considering its widespread availability and low immunogenicity human umbilical cord blood (HUCB) is a particularly attractive stem cell source. Our goal was to investigate the neurorestorative potential of cryopreserved HUCB mononuclear cells (MNC) after permanent middle cerebral artery occlusion (MCAO) in spontaneously hypertensive rats (SHR). Human umbilical cord blood MNC or vehicle solution was administered intravenously 24 hours after MCAO. Experimental groups were as follows: (1) quantitative polymerase chain reaction (PCR) of host-derived growth factors up to 48 hours after stroke; (2) immunohistochemical analysis of astroglial scarring; (3) magnetic resonance imaging (MRI) and weekly behavioral tests for 2 months after stroke. Long-term functional outcome and lesion development on MRI were not beneficially influenced by HUCB MNC therapy. Furthermore, HUCB MNC treatment did not change local growth factor levels and glial scarring extent. In summary, we could not demonstrate neurorestorative properties of HUCB MNC after stroke in SHR. Our results advise caution regarding a prompt translation of cord blood therapy into clinical stroke trials as long as deepened knowledge about its precise modes of action is missing.
Collapse
|
49
|
Mitkari B, Nitzsche F, Kerkelä E, Kuptsova K, Huttunen J, Nystedt J, Korhonen M, Jolkkonen J. Human bone marrow mesenchymal stem/stromal cells produce efficient localization in the brain and enhanced angiogenesis after intra-arterial delivery in rats with cerebral ischemia, but this is not translated to behavioral recovery. Behav Brain Res 2013; 259:50-9. [PMID: 24177208 DOI: 10.1016/j.bbr.2013.10.030] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 10/16/2013] [Accepted: 10/18/2013] [Indexed: 02/07/2023]
Abstract
Intravascular cell therapy is a promising approach for the treatment of stroke. However, high accumulation of cells to lungs and other filtering organs is a major concern after intravenous (i.v.) cell transplantation. This can be circumvented by intra-arterial (i.a.) cell infusion, which improves homing of cells to the injured brain. We studied the effect of i.a. delivery of human bone marrow-derived mesenchymal cells (BMMSCs) on behavioral and histological outcome in rats after middle cerebral artery occlusion (MCAO). Sixty male Wistar rats were subjected to transient MCAO (60 min) or sham-operation. BMMSCs (1×10(6)) were infused into the external carotid artery on postoperative day 2 or 7. Histology performed after a 42-day follow-up did not detect any human cells (MAB1281) in the ischemic brain. Endothelial cell staining with RECA-1 revealed a significant increase in the number of blood vessels in the perilesional cortex in MCAO rats treated with cells on postoperative day 7. Behavioral recovery as assessed in three tests, sticky label, cylinder and Montoya's staircase, was not improved by human BMMSCs during the follow-up. In conclusion, human BMMSCs did not improve functional recovery in MCAO rats despite effective initial homing to the ischemic hemisphere and enhanced angiogenesis, when strict behavioral tests not affected by repeated testing and compensation were utilized.
Collapse
Affiliation(s)
- Bhimashankar Mitkari
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Yliopistonranta 1 C, Kuopio, Finland
| | - Franziska Nitzsche
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Yliopistonranta 1 C, Kuopio, Finland
| | - Erja Kerkelä
- Finnish Red Cross Blood Service, Advanced Therapies and Product Development, Helsinki, Finland
| | - Kristina Kuptsova
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Yliopistonranta 1 C, Kuopio, Finland
| | - Joanna Huttunen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Johanna Nystedt
- Finnish Red Cross Blood Service, Advanced Therapies and Product Development, Helsinki, Finland
| | - Matti Korhonen
- Finnish Red Cross Blood Service, Advanced Therapies and Product Development, Helsinki, Finland
| | - Jukka Jolkkonen
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Yliopistonranta 1 C, Kuopio, Finland.
| |
Collapse
|
50
|
Affiliation(s)
- Sean I Savitz
- Department of Neurology, University of Texas Health Science Center, Houston, TX 77030, USA.
| |
Collapse
|