51
|
Mesenchymal Stromal Cell Therapy for Neonatal Hypoxic-Ischemic Encephalopathy. STEM CELLS IN CLINICAL APPLICATIONS 2017. [DOI: 10.1007/978-3-319-33720-3_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
52
|
Finn D, Roehr CC, Ryan CA, Dempsey EM. Optimising Intravenous Volume Resuscitation of the Newborn in the Delivery Room: Practical Considerations and Gaps in Knowledge. Neonatology 2017; 112:163-171. [PMID: 28571020 DOI: 10.1159/000475456] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 04/03/2017] [Indexed: 01/14/2023]
Abstract
Volume resuscitation (VR) for the treatment of newborn shock is a rare but potentially lifesaving intervention. Conducting clinical studies to assess the effectiveness of VR in the delivery room during newborn stabilization is challenging. We review the available literature and current management guidelines to determine which infants will benefit from VR, the frequency of VR, and the choice of agents used. In addition, the potential role for placental transfusion in the prevention of newborn shock is explored.
Collapse
Affiliation(s)
- Daragh Finn
- Department of Paediatrics and Child Health, Cork University Maternity Hospital and University College Cork, Cork, Ireland
| | | | | | | |
Collapse
|
53
|
Yıldız EP, Ekici B, Tatlı B. Neonatal hypoxic ischemic encephalopathy: an update on disease pathogenesis and treatment. Expert Rev Neurother 2016; 17:449-459. [PMID: 27830959 DOI: 10.1080/14737175.2017.1259567] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Hypoxic ischemic encephalopathy (HIE) is the most important reason for morbidity and mortality in term-born infants. Understanding pathophysiology of the brain damage is essential for the early detection of patients with high risk for HIE and development of strategies for their treatments. Areas covered: This review discusses pathophysiology of the neonatal HIE and its treatment options, including hypothermia, melatonin, allopurinol, topiramate, erythropoietin, N-acetylcyctein, magnesium sulphate and xenon. Expert commentary: Several clinical studies have been performed in order to decrease the risk of brain injury due to difficulties in the early diagnosis and treatment, and to develop strategies for better long-term outcomes. Although currently standard treatment methods include therapeutic hypothermia for neonates with moderate to severe HIE, new supportive options are needed to enhance neuroprotective effects of the hypothermia, which should aim to reduce production of the free radicals and to have anti-inflammatory and anti-apoptotic actions.
Collapse
Affiliation(s)
| | - Barış Ekici
- b Department of Pediatric Neurology , Liv Hospital , Istanbul , Turkey
| | - Burak Tatlı
- a Department of Pediatric Neurology , Istanbul University , Istanbul , Turkey
| |
Collapse
|
54
|
Kidani Y, Miki Y, Nomimura N, Minakawa S, Tanaka N, Miyoshi H, Wakabayashi K, Kudo Y. The therapeutic effect of CD133+ cells derived from human umbilical cord blood on neonatal mouse hypoxic-ischemic encephalopathy model. Life Sci 2016; 157:108-115. [DOI: 10.1016/j.lfs.2016.06.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 05/26/2016] [Accepted: 06/04/2016] [Indexed: 12/22/2022]
|
55
|
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
|
56
|
First Autologous Cord Blood Therapy for Pediatric Ischemic Stroke and Cerebral Palsy Caused by Cephalic Molding during Birth: Individual Treatment with Mononuclear Cells. Case Rep Transplant 2016; 2016:1717426. [PMID: 27239361 PMCID: PMC4867064 DOI: 10.1155/2016/1717426] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 04/12/2016] [Indexed: 12/05/2022] Open
Abstract
Intracranial laceration due to traumatic birth injury is an extremely rare event affecting approximately one newborn per a population of 4.5 million. However, depending on the mode of injury, the resulting brain damage may lead to lifelong sequelae, for example, cerebral palsy for which there is no cure at present. Here we report a rare case of neonatal arterial ischemic stroke and cerebral palsy caused by fetal traumatic molding and parietal depression of the head during delivery caused by functional cephalopelvic disproportion due to a “long pelvis.” This patient was treated by autologous cord blood mononuclear cells (45.8 mL, cryopreserved, TNC 2.53 × 10e8) with a remarkable recovery. Active rehabilitation was provided weekly. Follow-up examinations were at 3, 18, 34, and 57 months. Generous use of neonatal head MRI in case of molding, craniofacial deformity, and a sentinel event during parturition is advocated to enhance diagnosis of neonatal brain damage as a basis for fast and potentially causative treatment modalities including autologous cord blood transplantation in a timely manner.
Collapse
|
57
|
Abstract
Despite recent advances in neonatal intensive care medicine, neonatal brain injury resulting from intraventricular hemorrhage or hypoxic-ischemic encephalopathy remains a major cause of neonatal mortality and neurologic morbidities in survivors. Several studies have indicated that stem cell therapy is a promising novel therapy for neonatal brain injury resulting from these disorders. This review summarizes recent advances in stem cell research for treating neonatal brain injury due to intraventricular hemorrhage or hypoxic-ischemic encephalopathy with a particular focus on preclinical data, covering important issues for clinical translation such as optimal cell type, route, dose and timing of stem cell therapy, and translation of these preclinical results into a clinical trial.
Collapse
Affiliation(s)
- So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | |
Collapse
|
58
|
Aridas JDS, McDonald CA, Paton MCB, Yawno T, Sutherland AE, Nitsos I, Pham Y, Ditchfield M, Fahey MC, Wong F, Malhotra A, Castillo-Melendez M, Bhakoo K, Wallace EM, Jenkin G, Miller SL. Cord blood mononuclear cells prevent neuronal apoptosis in response to perinatal asphyxia in the newborn lamb. J Physiol 2015; 594:1421-35. [PMID: 26527561 DOI: 10.1113/jp271104] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/23/2015] [Indexed: 11/08/2022] Open
Abstract
Perinatal asphyxia is a significant cause of death or long-term neurodevelopmental impairment. Hypothermia, currently the only effective treatment, leads to modest improvements, but new therapeutic strategies are required. Umbilical cord blood (UCB) mononuclear cells have potent anti-inflammatory properties and may reduce neuropathology. This study examined whether autologous UCB mononuclear cells were neuroprotective when administered to newborn lambs at 12 h after birth asphyxia. At caesarean section, birth asphyxia was induced by clamping the umbilical cord until mean arterial blood pressure decreased to 18-20 mmHg. Asphyxia (n = 20) or control (n = 11) lambs were resuscitated and maintained, with magnetic resonance spectroscropy (MRS) performed at 12 and 72 h, and were then killed at 72 h. Cord blood was collected once the cord was clamped, and mononuclear cells were isolated and labelled fluorescently and administered to control (n = 3) or asphyxia (n = 8) lambs. Asphyxia induced a significant increase in cellular apoptosis (caspase-3 immunopositive) within all brain regions examined, including cortex, hippocampus, thalamus, striatum and subcortical white matter (P < 0.01 vs. control). Additionally, asphyxia induced significant and widespread astrogliosis and increased inflammatory cells (activated microglia and macrophages). The administration of UCB mononuclear cells (asphyxia+UCB) significantly decreased neuronal apoptosis, astrogliosis and inflammation (P < 0.05 vs. asphyxia alone). Asphyxia+UCB lambs also demonstrated decreased brain metabolites lactate:choline (P = 0.01) and lactate:N-acetylaspartate (P < 0.01) from 12 to 72 h, detected using MRS. Autologous UCB mononuclear cell treatment restores normal brain metabolism following perinatal asphyxia, and reduces brain inflammation, astrogliosis and neuronal apoptosis, supporting its use as a neuroprotective therapy following asphyxia.
Collapse
Affiliation(s)
- James D S Aridas
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Courtney A McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Madison C B Paton
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Tamara Yawno
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Amy E Sutherland
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Ilias Nitsos
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Yen Pham
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Michael Ditchfield
- Monash Children's, Monash Health, and Department of Paediatrics, Monash University, Clayton, Victoria, Australia.,Diagnostic Imaging, Monash Health, Clayton, Victoria, Australia
| | - Michael C Fahey
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Monash Children's, Monash Health, and Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Flora Wong
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Monash Children's, Monash Health, and Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Atul Malhotra
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Monash Children's, Monash Health, and Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Margie Castillo-Melendez
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Kishore Bhakoo
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A*STAR), Biopolis Way, Singapore
| | - Euan M Wallace
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
59
|
Ghaffaripour HA, Jalali M, Nikravesh MR, Seghatoleslam M, Sanchooli J. Neuronal cell reconstruction with umbilical cord blood cells in the brain hypoxia-ischemia. IRANIAN BIOMEDICAL JOURNAL 2015; 19:29-34. [PMID: 25605487 PMCID: PMC4322230 DOI: 10.6091/ibj.1376.2015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background: Brain hypoxia-ischemia is a human neonatal injury that is considered a candidate for stem cell therapy. Methods: The possible therapeutic potential of human umbilical cord blood (HUCB) stem cells was evaluated in 14-day-old rats subjected to the right common carotid occlusion, a model of neonatal brain hypoxia-ischemia. Seven days after hypoxia-ischemia, rats received either saline solution or 4 × 105 HUCB cells i.v. Rats in control group did not receive any injection. After two weeks, rats were assessed using two motor tests. Subsequently, rats were scarified for histological and immunohistochemical analyses. Results: Our immunohistochemical findings demonstrated selective migration of the injected HUCB cells to the ischemic area as well as reduction in infarct volume. Seven days after surgery, we found significant recovery in the behavioral performance in the test group (12.7 +/- 0.3) compared to the sham group (10.0 +/-0.05), a trend which continued to day 14 (15.3 ± 0.3 vs. 11.9 ± 0.5, P<0.05). Postural and motor asymmetries at days 7 and 14 in the test group showed a significant decrease in the percentage of right turns in comparison to the sham group (75% and 59% vs. 97% and 96%, P<0.05). Conclusion: The results show the potential of HUCB stem cells in reduction of neurologic deficits associated with neonatal hypoxia-ischemia.
Collapse
Affiliation(s)
| | - Mehdi Jalali
- Dept. of Anatomy and Cell Biology, Medical School, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Nikravesh
- Dept. of Anatomy and Cell Biology, Medical School, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Masoumeh Seghatoleslam
- Dept. of Anatomy and Cell Biology, Medical School, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Sanchooli
- Dept. of Biochemistry and Immunology, Medical School, Zabol Medical Science University, Zabol, Iran
| |
Collapse
|
60
|
[Perinatal neuroprotection in 2015]. Arch Pediatr 2015; 22:1005-7. [PMID: 26382640 DOI: 10.1016/j.arcped.2015.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/10/2015] [Accepted: 07/02/2015] [Indexed: 11/20/2022]
|
61
|
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
|
62
|
Dixon BJ, Reis C, Ho WM, Tang J, Zhang JH. Neuroprotective Strategies after Neonatal Hypoxic Ischemic Encephalopathy. Int J Mol Sci 2015; 16:22368-401. [PMID: 26389893 PMCID: PMC4613313 DOI: 10.3390/ijms160922368] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/31/2015] [Accepted: 09/06/2015] [Indexed: 12/21/2022] Open
Abstract
Neonatal hypoxic ischemic encephalopathy (HIE) is a devastating disease that primarily causes neuronal and white matter injury and is among the leading cause of death among infants. Currently there are no well-established treatments; thus, it is important to understand the pathophysiology of the disease and elucidate complications that are creating a gap between basic science and clinical translation. In the development of neuroprotective strategies and translation of experimental results in HIE, there are many limitations and challenges to master based on an appropriate study design, drug delivery properties, dosage, and use in neonates. We will identify understudied targets after HIE, as well as neuroprotective molecules that bring hope to future treatments such as melatonin, topiramate, xenon, interferon-beta, stem cell transplantation. This review will also discuss some of the most recent trials being conducted in the clinical setting and evaluate what directions are needed in the future.
Collapse
Affiliation(s)
- Brandon J Dixon
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| | - Cesar Reis
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
| | - Wing Mann Ho
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
- Department of Neurosurgery, Medical University Innsbruck, Tyrol 6020, Austria.
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
- Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| |
Collapse
|
63
|
Abstract
Stem cell transplantation (SCT) is an established first-line or adjunctive therapy for a variety of neonatal and adult diseases. New evidence in preclinical models as well as a few human studies show the potential utility of SCT in neuroprotection and in the modulation of inflammatory injury in at risk-neonates. This review briefly summarizes current understanding of human stem cell biology during ontogeny and present recent evidence supporting SCT as a viable approach for postinsult neonatal injury.
Collapse
Affiliation(s)
- Momoko Yoshimoto
- Assistant Research Professor, Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, 1044W Walnut Street R4-W116, Indianapolis, IN 46202, Tel: 317-278-0598
| | - Joyce M Koenig
- Pediatrics, E Doisy Research Center, Saint Louis University School of Medicine, 1100 South Grand Boulevard, St Louis, MO 63104, USA; Molecular Microbiology & Immunology, E Doisy Research Center, Saint Louis University School of Medicine, 1100 South Grand Boulevard, St Louis, MO 63106, USA.
| |
Collapse
|
64
|
Titomanlio L, Fernández-López D, Manganozzi L, Moretti R, Vexler ZS, Gressens P. Pathophysiology and neuroprotection of global and focal perinatal brain injury: lessons from animal models. Pediatr Neurol 2015; 52:566-584. [PMID: 26002050 PMCID: PMC4720385 DOI: 10.1016/j.pediatrneurol.2015.01.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 01/16/2015] [Accepted: 01/24/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Arterial ischemic stroke occurs more frequently in term newborns than in the elderly, and brain immaturity affects mechanisms of ischemic injury and recovery. The susceptibility to injury of the brain was assumed to be lower in the perinatal period as compared with childhood. This concept was recently challenged by clinical studies showing marked motor disabilities after stroke in neonates, with the severity of motor and cortical sensory deficits similar in both perinatal and childhood ischemic stroke. Our understanding of the triggers and the pathophysiological mechanisms of perinatal stroke has greatly improved in recent years, but many factors remain incompletely understood. METHODS In this review, we focus on the pathophysiology of perinatal stroke and on therapeutic strategies that can protect the immature brain from the consequences of stroke by targeting inflammation and brain microenvironment. RESULTS Studies in neonatal rodent models of cerebral ischemia have suggested a potential role for soluble inflammatory molecules as important modulators of injury and recovery. A great effort is underway to investigate neuroprotective molecules based on our increasing understanding of the pathophysiology. CONCLUSION In this review, we provide a comprehensive summary of new insights concerning pathophysiology of focal and global perinatal brain injury and their implications for new therapeutic approaches.
Collapse
Affiliation(s)
- Luigi Titomanlio
- Pediatric Emergency Department, APHP, Robert Debré Hospital, Paris, France
- Inserm, U1141, F-75019 Paris, France
| | - David Fernández-López
- Department of Neurology, University of California San Francisco, San Francisco, CA, 94158-0663, USA
| | - Lucilla Manganozzi
- Pediatric Emergency Department, APHP, Robert Debré Hospital, Paris, France
- Inserm, U1141, F-75019 Paris, France
| | | | - Zinaida S. Vexler
- Department of Neurology, University of California San Francisco, San Francisco, CA, 94158-0663, USA
| | - Pierre Gressens
- Inserm, U1141, F-75019 Paris, France
- Univ Paris Diderot, Sorbonne Paris Cité, UMRS 676, F-75019 Paris, France
- PremUP, Paris, France
- Centre for the Developing Brain, King’s College, St Thomas’ Campus, London SE1 7EH, UK
| |
Collapse
|
65
|
Ohshima M, Taguchi A, Tsuda H, Sato Y, Yamahara K, Harada-Shiba M, Miyazato M, Ikeda T, Iida H, Tsuji M. Intraperitoneal and intravenous deliveries are not comparable in terms of drug efficacy and cell distribution in neonatal mice with hypoxia-ischemia. Brain Dev 2015; 37:376-86. [PMID: 25034178 DOI: 10.1016/j.braindev.2014.06.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 06/23/2014] [Accepted: 06/23/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND PURPOSE Most therapeutic agents are administered intravenously (IV) in clinical settings and intraperitoneally (IP) in preclinical studies with neonatal rodents; however, it remains unclear whether intraperitoneal (IP) injection is truly an acceptable alternative for intravenous (IV) injection in preclinical studies. The objective of our study is to clarify the differences in the therapeutic effects of drugs and in the distribution of infused cells after an IP or IV injection in animals with brain injury. METHODS Dexamethasone or MK-801, an N-methyl-d-aspartate receptor antagonist was administered either IP or IV in a mouse model of neonatal hypoxic-ischemic encephalopathy. Green fluorescent protein-expressing mesenchymal stem cells (MSCs) or mononuclear cells (MNCs) were injected IP or IV in the mouse model. Two hours and 24h after the administration of the cells, we investigated the cell distributions by immunohistochemical staining. We also investigated distribution of IV administered MNCs labeled with 2-[18F]fluoro-2-deoxy-d-glucose in a juvenile primate, a macaque with stroke 1h after the administration. RESULTS IP and IV administration of dexamethasone attenuated the brain injury to a similar degree. IP administration of MK-801 attenuated brain injury, whereas IV administration of MK-801 did not. The IV group showed a significantly greater number of infused cells in the lungs and brains in the MSC cohort and in the spleen, liver, and lung in the MNC cohort compared to the IP group. In the macaque, MNCs were detected in the spleen and liver in large amounts, but not in the brain and lungs. CONCLUSIONS This study demonstrated that the administration route influences the effects of drugs and cell distribution. Therefore, a preclinical study may need to be performed using the optimal administration route used in a clinical setting.
Collapse
Affiliation(s)
- Makiko Ohshima
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Akihiko Taguchi
- Department of Regenerative Medicine Research, Institute of Biomedical Research Innovation, Kobe, Hyogo, Japan
| | - Hidetoshi Tsuda
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Kenichi Yamahara
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Mariko Harada-Shiba
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Mikiya Miyazato
- Department of Biochemistry, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Tomoaki Ikeda
- Department of Obstetrics and Gynecology, Mie University School of Medicine, Tsu, Mie, Japan
| | - Hidehiro Iida
- Department of Investigative Radiology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Masahiro Tsuji
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan.
| |
Collapse
|
66
|
Merchant NM, Azzopardi DV, Edwards AD. Neonatal hypoxic ischaemic encephalopathy: current and future treatment options. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1021776] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
67
|
Chen LX, Ma SM, Zhang P, Fan ZC, Xiong M, Cheng GQ, Yang Y, Qiu ZL, Zhou WH, Li J. Neuroprotective effects of oligodendrocyte progenitor cell transplantation in premature rat brain following hypoxic-ischemic injury. PLoS One 2015; 10:e0115997. [PMID: 25790286 PMCID: PMC4366232 DOI: 10.1371/journal.pone.0115997] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/03/2014] [Indexed: 01/19/2023] Open
Abstract
Periventricular leukomalacia (PVL) is a common ischemic brain injury in premature infants for which there is no effective treatment. The objective of this study was to determine whether transplanted mouse oligodendrocyte progenitor cells (OPCs) have neuroprotective effects in a rat model of PVL. Hypoxia-ischemia (HI) was induced in 3-day-old rat pups by left carotid artery ligation, followed by exposure to 6% oxygen for 2.5 h. Animals were assigned to OPC transplantation or sham control groups and injected with OPCs or PBS, respectively, and sacrificed up to 6 weeks later for immunohistochemical analysis to investigate the survival and differentiation of transplanted OPCs. Apoptosis was evaluated by double immunolabeling of brain sections for caspase-3 and neuronal nuclei (NeuN), while proliferation was assessed using a combination of anti-Nestin and -bromodeoxyuridine antibodies. The expression of brain-derived neurotrophic factor (BDNF) and Bcl-2 was examined 7 days after OPC transplantation. The Morris water maze was used to test spatial learning and memory. The results showed that transplanted OPCs survived and formed a myelin sheath, and stimulated BDNF and Bcl-2 expression and the proliferation of neural stem cells (NSC), while inhibiting HI-induced neuronal apoptosis relative to control animals. Moreover, deficits in spatial learning and memory resulting from HI were improved by OPC transplantation. These results demonstrate an important neuroprotective role for OPCs that can potentially be exploited in cell-based therapeutic approaches to minimize HI-induced brain injury.
Collapse
Affiliation(s)
- Long-Xia Chen
- Key Laboratory of Birth Defect, Children’s Hospital of Fudan University, Shanghai, China
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
| | - Si-Min Ma
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, 201102, China
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
| | - Peng Zhang
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
| | - Zi-Chuan Fan
- Key Laboratory of Birth Defect, Children’s Hospital of Fudan University, Shanghai, China
| | - Man Xiong
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
| | - Guo-Qiang Cheng
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, 201102, China
| | - Yi Yang
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
| | - Zi-Long Qiu
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wen-Hao Zhou
- Department of Neonatology, Children’s Hospital of Fudan University, Shanghai, 201102, China
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children’s Hospital of Fudan University, Shanghai, China
- * E-mail: (WZ); (JL)
| | - Jin Li
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- * E-mail: (WZ); (JL)
| |
Collapse
|
68
|
Drobyshevsky A, Cotten CM, Shi Z, Luo K, Jiang R, Derrick M, Tracy ET, Gentry T, Goldberg RN, Kurtzberg J, Tan S. Human Umbilical Cord Blood Cells Ameliorate Motor Deficits in Rabbits in a Cerebral Palsy Model. Dev Neurosci 2015; 37:349-62. [PMID: 25791742 DOI: 10.1159/000374107] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 01/09/2015] [Indexed: 11/19/2022] Open
Abstract
Cerebral palsy (CP) has a significant impact on both patients and society, but therapy is limited. Human umbilical cord blood cells (HUCBC), containing various stem and progenitor cells, have been used to treat various brain genetic conditions. In small animal experiments, HUCBC have improved outcomes after hypoxic-ischemic (HI) injury. Clinical trials using HUCBC are underway, testing feasibility, safety and efficacy for neonatal injury as well as CP. We tested HUCBC therapy in a validated rabbit model of CP after acute changes secondary to HI injury had subsided. Following uterine ischemia at 70% gestation, we infused HUCBC into newborn rabbit kits with either mild or severe neurobehavioral changes. Infusion of high-dose HUCBC (5 × 10(6) cells) dramatically altered the natural history of the injury, alleviating the abnormal phenotype including posture, righting reflex, locomotion, tone, and dystonia. Half the high dose showed lesser but still significant improvement. The swimming test, however, showed that joint function did not restore to naïve control function in either group. Tracing HUCBC with either MRI biomarkers or PCR for human DNA found little penetration of HUCBC in the newborn brain in the immediate newborn period, suggesting that the beneficial effects were not due to cellular integration or direct proliferative effects but rather to paracrine signaling. This is the first study to show that HUCBC improve motor performance in a dose-dependent manner, perhaps by improving compensatory repair processes.
Collapse
|
69
|
Zhang C, Huang L, Gu J, Zhou X. Therapy for Cerebral Palsy by Human Umbilical Cord Blood Mesenchymal Stem Cells Transplantation Combined With Basic Rehabilitation Treatment: A Case Report. Glob Pediatr Health 2015; 2:2333794X15574091. [PMID: 27335947 PMCID: PMC4784638 DOI: 10.1177/2333794x15574091] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background. Cerebral palsy (CP) is the most common cause leading to childhood disability. Human umbilical cord blood mesenchymal stem cells (hUCB-MSCs) transplantation is a promising alternative considering the safety and efficacy in current reports. This report represents a case of hUCB-MSCs transplantation combined with basic rehabilitation treatment beginning as early as age 6 months with follow-up as long as 5 years. Methods. A 6-year-old female patient was diagnosed with CP at age 6 months. The patient accepted 4 infusions of intravenous hUCB-MSCs in each course and received 4 courses of transplantation totally. A series of assessments were performed before the first transplantation, including laboratory tests, CDCC Infant Mental Development Scale, and Gross Motor Function Measure-88 (GMFM-88). Then annual assessments using the GMFM-88, Ashworth spasm assessment, and comprehensive function assessment scale were made in addition to the annual laboratory tests. In addition, electroencephalography and brain magnetic resonance imaging were conducted before transplantation and in the follow-up phase. Rehabilitation and safety follow-up have been ongoing for 5 years up to date. Results. There was no complaint about adverse effects during hospitalization or postoperative follow-up. Motor function recovered to normal level according to the evaluation of scales. Language function improved significantly. Linguistic rehabilitation therapy was enhanced for further improvement. Conclusions. The clinical application of hUC-MSCs combined with basic rehabilitation treatment was effective and safe for improving motor and comprehensive function in a patient with CP.
Collapse
Affiliation(s)
- Che Zhang
- Taihe Hospital Affiliated to Hubei Medical College, Shiyan, China
| | - Li Huang
- Taihe Hospital Affiliated to Hubei Medical College, Shiyan, China
| | - Jiaowei Gu
- Taihe Hospital Affiliated to Hubei Medical College, Shiyan, China
| | - Xihui Zhou
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
70
|
Hattori T, Sato Y, Kondo T, Ichinohashi Y, Sugiyama Y, Yamamoto M, Kotani T, Hirata H, Hirakawa A, Suzuki S, Tsuji M, Ikeda T, Nakanishi K, Kojima S, Blomgren K, Hayakawa M. Administration of umbilical cord blood cells transiently decreased hypoxic-ischemic brain injury in neonatal rats. Dev Neurosci 2015; 37:95-104. [PMID: 25720519 DOI: 10.1159/000368396] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 09/12/2014] [Indexed: 11/19/2022] Open
Abstract
This study aimed to investigate whether the administration of mononuclear cells derived from human umbilical cord blood cells (UCBCs) could ameliorate hypoxic-ischemic brain injury in a neonatal rat model. The left carotid arteries of 7-day-old rats were ligated, and the rats were then exposed to 8% oxygen for 60 min. Mononuclear cells derived from UCBCs using the Ficoll-Hypaque technique were injected intraperitoneally 6 h after the insult (1.0 × 10(7) cells). Twenty-four hours after the insult, the number of cells positive for the oxidative stress markers 4-hydroxy-2-nonenal and nitrotyrosine, in the dentate gyrus of the hippocampus in the UCBC-treated group, decreased by 36 and 42%, respectively, compared with those in the control group. In addition, the number of cells positive for the apoptosis markers active caspase-3 and apoptosis-inducing factor decreased by 53 and 58%, respectively. The number of activated microglia (ED1-positive cells) was 51% lower in the UCBC group compared with the control group. In a gait analysis performed 2 weeks after the insult, there were no significant differences among the sham-operated, control and UCBC groups. An active avoidance test using a shuttle box the following week also revealed no significant differences among the groups. Neither the volumes of the hippocampi, corpus callosum and cortices nor the numbers of neurons in the hippocampus were different between the UCBC and control groups. In summary, a single intraperitoneal injection of UCBC-derived mononuclear cells 6 h after an ischemic insult was associated with a transient reduction in numbers of apoptosis and oxidative stress marker-positive cells, but it did not induce long-term morphological or functional protection. Repeated administration or a combination treatment may be required to achieve sustained protection.
Collapse
Affiliation(s)
- Tetsuo Hattori
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Fan HC, Ho LI, Chi CS, Cheng SN, Juan CJ, Chiang KL, Lin SZ, Harn HJ. Current proceedings of cerebral palsy. Cell Transplant 2015; 24:471-85. [PMID: 25706819 DOI: 10.3727/096368915x686931] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cerebral palsy (CP) is a complicated disease with varying causes and outcomes. It has created significant burden to both affected families and societies, not to mention the quality of life of the patients themselves. There is no cure for the disease; therefore, development of effective therapeutic strategies is in great demand. Recent advances in regenerative medicine suggest that the transplantation of stem cells, including embryonic stem cells, neural stem cells, bone marrow mesenchymal stem cells, induced pluripotent stem cells, umbilical cord blood cells, and human embryonic germ cells, focusing on the root of the problem, may provide the possibility of developing a complete cure in treating CP. However, safety is the first factor to be considered because some stem cells may cause tumorigenesis. Additionally, more preclinical and clinical studies are needed to determine the type of cells, route of delivery, cell dose, timing of transplantation, and combinatorial strategies to achieve an optimal outcome.
Collapse
Affiliation(s)
- Hueng-Chuen Fan
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Ding HF, Zhang H, Ding HF, Li D, Yi XH, Gao XY, Mou WW, Ju XL. Therapeutic effect of placenta-derived mesenchymal stem cells on hypoxic-ischemic brain damage in rats. World J Pediatr 2015; 11:74-82. [PMID: 25447631 DOI: 10.1007/s12519-014-0531-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 04/09/2014] [Indexed: 12/24/2022]
Abstract
BACKGROUND Oxidative stress is involved in the development of hypoxic-ischemic brain damage (HIBD). In this study, we investigated the therapeutic effects of placenta-derived mesenchymal stem cells (PD-MSCs) and explored the NF-E2-related factor-2/heme oxygenase-1 (Nrf2/HO-1) signaling pathway in treating HIBD. METHODS P7 rats were subjected to hypoxic-ischemic brain injury and randomly divided into four groups (control, HIBD, HIBD+PD-MSCs, and HIBD+fibroblasts). Forty-eight hours after the induction of HIBD, 5×10(5) of PD-MSCs were injected into cerebral tissue in the HIBD+PD-MSCs group, while the same dose of fibroblasts were injected in the HIBD+fibroblasts group. Morris Water Maze, gross and pathological changes were tested at P28. The level of malondialdehyde (MDA) was detected in rats' hippocampus. RT-PCR and western blot analysis were used to evaluate the changes of Nrf2/HO-1. RESULTS The HIBD group showed significantly longer escape latency and a lower frequency of original platform crossing in the Morris Water Maze compared with the control group. Rats receiving PD-MSCs showed significant improvement of HIBD. The pathological changes were evident after HIBD, but ameliorated in the PD-MSCs group. Compared with the control group, HO-1 and Nrf2 were up-regulated at gene and protein levels in the HI brain, beginning at 6 hours and peaking at 48 hours (P<0.05). The expression of HO-1 and Nrf2 in the PD-MSCs treatment group was more pronounced than in the HIBD group (P<0.01). PD-MSCs also decreased MDA production in the brain tissue. CONCLUSION These results demonstrate that PD-MSCs have neuroprotective effect during the treatment of HIBD and that the mechanism may be partly due to alleviating oxidative stress.
Collapse
Affiliation(s)
- Hong-Fang Ding
- Department of Pediatrics, Shengli Oil Field Central Hospital, Dongying, 257034, China
| | | | | | | | | | | | | | | |
Collapse
|
73
|
|
74
|
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
|
75
|
Li Y, Hua X, Hua F, Mao W, Wan L, Li S. Are bone marrow regenerative cells ideal seed cells for the treatment of cerebral ischemia? Neural Regen Res 2014; 8:1201-9. [PMID: 25206414 PMCID: PMC4107607 DOI: 10.3969/j.issn.1673-5374.2013.13.005] [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: 12/30/2012] [Accepted: 03/27/2013] [Indexed: 01/16/2023] Open
Abstract
Bone marrow cells for the treatment of ischemic brain injury may depend on the secretion of a large number of neurotrophic factors. Bone marrow regenerative cells are capable of increasing the secretion of neurotrophic factors. In this study, after tail vein injection of 5-fluorouracil for 7 days, bone marrow cells and bone marrow regenerative cells were isolated from the tibias and femurs of rats, and then administered intravenously via the tail vein after focal cerebral ischemia. Immunohistological staining and reverse transcription-PCR detection showed that transplanted bone marrow cells and bone marrow regenerative cells could migrate and survive in the ischemic regions, such as the cortical and striatal infarction zone. These cells promote vascular endothelial cell growth factor mRNA expression in the ischemic marginal zone surrounding the ischemic penumbra of the cortical and striatal infarction zone, and have great advantages in promoting the recovery of neurological function, reducing infarct size and promoting angiogenesis. Bone marrow regenerative cells exhibited stronger neuroprotective effects than bone marrow cells. Our experimental findings indicate that bone marrow regenerative cells are preferable over bone marrow cells for cell therapy for neural regeneration after cerebral ischemia. Their neuroprotective effect is largely due to their ability to induce the secretion of factors that promote vascular regeneration, such as vascular endothelial growth factor.
Collapse
Affiliation(s)
- Yi Li
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xuming Hua
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Fang Hua
- Department of Emergency Medicine, Brain Research Laboratory, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Wenwei Mao
- Laboratory of Microbiology and Biochemical Pharmaceutics, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Liang Wan
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Shiting Li
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
76
|
Abstract
Perinatal brain injuries are a leading cause of cerebral palsy worldwide. The potential of stem cell therapy to prevent or reduce these impairments has been widely discussed within the medical and scientific communities and an increasing amount of research is being conducted in this field. Animal studies support the idea that a number of stem cells types, including cord blood and mesenchymal stem cells have a neuroprotective effect in neonatal hypoxia-ischemia. Both these cell types are readily available in a clinical setting. The mechanisms of action appear to be diverse, including immunomodulation, activation of endogenous stem cells, release of growth factors, and anti-apoptotic effects. Here, we review the different types of stem cells and progenitor cells that are potential candidates for therapeutic strategies in perinatal brain injuries, and summarize recent preclinical and clinical studies.
Collapse
Affiliation(s)
- Andre W Phillips
- The Hugo W. Moser Research Institute at Kennedy Krieger Institute Johns Hopkins University, Baltimore, Maryland, USA ; Department of Neurology Johns Hopkins University, Baltimore, Maryland, USA
| | | | | |
Collapse
|
77
|
Wang F, Shen Y, Tsuru E, Yamashita T, Baba N, Tsuda M, Maeda N, Sagara Y. Syngeneic transplantation of newborn splenocytes in a murine model of neonatal ischemia-reperfusion brain injury. J Matern Fetal Neonatal Med 2014; 28:842-7. [PMID: 24939627 DOI: 10.3109/14767058.2014.935327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Neonatal hypoxic-ischemic encephalopathy (HIE) is caused by brain injury that occurs in a developing fetus or infant. Stem cell transplantation can reportedly induce functional recovery in animal models of HIE. Murine neonatal splenocytes are enriched with immature blood stem cells and are used for the investigation of murine models of syngeneic transplantation. The aim of this study was to investigate the therapeutic potential of newborn splenocytes in a murine model of neonatal ischemia-reperfusion brain injury. METHODS C57BL/6N mice (postnatal day 7) underwent right common carotid artery occlusion with an aneurysm clip. Following hypoxic exposure, reperfusion was achieved by unclamping the artery. Newborn splenocytes were transplanted intravenously at 3 weeks after injury. RESULTS The splenocytes transplanted group tended to show an improvement in behavioral tests, but it was not significantly different compared with the control groups. The transplanted cells were localized in various organs including injured brain tissue over 3 weeks. In the penumbra region of the brain, vascular endothelial growth factor (VEGF) expression was upregulated after transplantation. CONCLUSIONS These results showed that syngeneic transplantation of newborn splenocytes achieved the long-term survival of the grafts and exerted influence the microenvironment in the injured brains of mice.
Collapse
Affiliation(s)
- Feifei Wang
- Center for Innovative and Translational Medicine, Kochi University Medical School , Nankoku, Kochi , Japan
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Donega V, Nijboer CH, van Tilborg G, Dijkhuizen RM, Kavelaars A, Heijnen CJ. Intranasally administered mesenchymal stem cells promote a regenerative niche for repair of neonatal ischemic brain injury. Exp Neurol 2014; 261:53-64. [PMID: 24945601 DOI: 10.1016/j.expneurol.2014.06.009] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 05/30/2014] [Accepted: 06/09/2014] [Indexed: 12/15/2022]
Abstract
Previous work from our group has shown that intranasal MSC-treatment decreases lesion volume and improves motor and cognitive behavior after hypoxic-ischemic (HI) brain damage in neonatal mice. Our aim was to determine the kinetics of MSC migration after intranasal administration, and the early effects of MSCs on neurogenic processes and gliosis at the lesion site. HI brain injury was induced in 9-day-old mice and MSCs were administered intranasally at 10days post-HI. The kinetics of MSC migration were investigated by immunofluorescence and MRI analysis. BDNF and NGF gene expression was determined by qPCR analysis following MSC co-culture with HI brain extract. Nestin, Doublecortin, NeuN, GFAP, Iba-1 and M1/M2 phenotypic expression was assessed over time. MRI and immunohistochemistry analyses showed that MSCs reach the lesion site already within 2h after intranasal administration. At 12h after administration the number of MSCs at the lesion site peaks and decreases significantly at 72h. The number of DCX(+) cells increased 1 to 3days after MSC administration in the SVZ. At the lesion, GFAP(+)/nestin(+) and DCX(+) expression increased 3 to 5days after MSC-treatment. The number of NeuN(+) cells increased within 5days, leading to a dramatic regeneration of the somatosensory cortex and hippocampus at 18days after intranasal MSC administration. Interestingly, MSCs expressed significantly more BDNF gene when exposed to HI brain extract in vitro. Furthermore, MSC-treatment resulted in the resolution of the glial scar surrounding the lesion, represented by a decrease in reactive astrocytes and microglia and polarization of microglia towards the M2 phenotype. In view of the current lack of therapeutic strategies, we propose that intranasal MSC administration is a powerful therapeutic option through its functional repair of the lesion represented by regeneration of the cortical and hippocampal structure and decrease of gliosis.
Collapse
Affiliation(s)
- Vanessa Donega
- Lab. of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cora H Nijboer
- Lab. of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Geralda van Tilborg
- Biomedical MR Imaging and Spectroscopy Group, Image Sciences Institute, University Medical Center Utrecht, The Netherlands
| | - Rick M Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Image Sciences Institute, University Medical Center Utrecht, The Netherlands
| | - Annemieke Kavelaars
- Lab. of Neuroimmunology, Department of Symptom Research, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Cobi J Heijnen
- Lab. of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands; Lab. of Neuroimmunology, Department of Symptom Research, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
79
|
Cotten CM, Murtha AP, Goldberg RN, Grotegut CA, Smith PB, Goldstein RF, Fisher KA, Gustafson KE, Waters-Pick B, Swamy GK, Rattray B, Tan S, Kurtzberg J. Feasibility of autologous cord blood cells for infants with hypoxic-ischemic encephalopathy. J Pediatr 2014; 164:973-979.e1. [PMID: 24388332 PMCID: PMC3992180 DOI: 10.1016/j.jpeds.2013.11.036] [Citation(s) in RCA: 223] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 10/25/2013] [Accepted: 11/14/2013] [Indexed: 10/25/2022]
Abstract
OBJECTIVE To assess feasibility and safety of providing autologous umbilical cord blood (UCB) cells to neonates with hypoxic-ischemic encephalopathy (HIE). STUDY DESIGN We enrolled infants in the intensive care nursery who were cooled for HIE and had available UCB in an open-label study of non-cyropreserved autologous volume- and red blood cell-reduced UCB cells (up to 4 doses adjusted for volume and red blood cell content, 1-5 × 10(7) cells/dose). We recorded UCB collection and cell infusion characteristics, and pre- and post-infusion vital signs. As exploratory analyses, we compared cell recipients' hospital outcomes (mortality, oral feeds at discharge) and 1-year survival with Bayley Scales of Infant and Toddler Development, 3rd edition scores ≥85 in 3 domains (cognitive, language, and motor development) with cooled infants who did not have available cells. RESULTS Twenty-three infants were cooled and received cells. Median collection and infusion volumes were 36 and 4.3 mL. Vital signs including oxygen saturation were similar before and after infusions in the first 48 postnatal hours. Cell recipients and concurrent cooled infants had similar hospital outcomes. Thirteen of 18 (74%) cell recipients and 19 of 46 (41%) concurrent cooled infants with known 1-year outcomes survived with scores >85. CONCLUSIONS Collection, preparation, and infusion of fresh autologous UCB cells for use in infants with HIE is feasible. A randomized double-blind study is needed.
Collapse
MESH Headings
- Child, Preschool
- Combined Modality Therapy
- Cord Blood Stem Cell Transplantation/methods
- Developmental Disabilities/diagnosis
- Developmental Disabilities/etiology
- Feasibility Studies
- Female
- Follow-Up Studies
- Humans
- Hypothermia, Induced
- Hypoxia-Ischemia, Brain/complications
- Hypoxia-Ischemia, Brain/mortality
- Hypoxia-Ischemia, Brain/surgery
- Hypoxia-Ischemia, Brain/therapy
- Infant
- Infant, Newborn
- Infant, Premature
- Infant, Premature, Diseases/mortality
- Infant, Premature, Diseases/surgery
- Infant, Premature, Diseases/therapy
- Male
- Pilot Projects
- Severity of Illness Index
- Transplantation, Autologous/methods
- Treatment Outcome
Collapse
Affiliation(s)
| | - Amy P Murtha
- Department of Obstetrics and Gynecology, Duke University, Durham, NC
| | | | - Chad A Grotegut
- Department of Obstetrics and Gynecology, Duke University, Durham, NC
| | - P Brian Smith
- Department of Pediatrics, Duke University, Durham, NC
| | | | | | - Kathryn E Gustafson
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC
| | | | - Geeta K Swamy
- Department of Obstetrics and Gynecology, Duke University, Durham, NC
| | | | - Siddhartha Tan
- Department of Pediatrics, NorthShore University Health System and University of Chicago Pritzker School of Medicine, Chicago, IL
| | - Joanne Kurtzberg
- Robertson Cell and Translational Therapy Program, Duke University, Durham, NC
| |
Collapse
|
80
|
Human umbilical cord blood mesenchymal stem cell transplantation suppresses inflammatory responses and neuronal apoptosis during early stage of focal cerebral ischemia in rabbits. Acta Pharmacol Sin 2014; 35:585-91. [PMID: 24727940 DOI: 10.1038/aps.2014.9] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 02/12/2014] [Indexed: 02/08/2023] Open
Abstract
AIM Human umbilical cord blood mesenchymal stem cells (hUCB-MSCs) have been shown to ameliorate cerebral ischemia in animal models. In this study we investigated the effects of hUCB-MSCs on inflammatory responses and neuronal apoptosis during the early stage of focal cerebral ischemia in rabbits. METHODS Focal cerebral ischemia was induced in male New Zealand rabbits by occlusion of MCA for 2 h. The blood samples were collected at different time points prior and during MCAO-reperfusion. The animals were euthanized 3 d after MCAO, and the protein levels of IL-1β, IL-6, IL-10 and TNF-α in the serum and peri-ischemic brain tissues were detected using Western blot and ELISA, respectively. Inflammatory cell infiltration, neuronal apoptosis and neuronal density were measured morphologically. hUCB-MSCs (5 × 10(6)) were iv injected a few minutes after MCAO. RESULTS The serum levels of IL-1β, IL-6 and TNF-α were rapidly increased, and peaked at 2 h after the start of MCAO. hUCB-MSC transplantation markedly and progressively suppressed the ischemia-induced increases of serum IL-1β, IL-6 and TNF-α levels within 6 h MCAO-reperfusion. Focal cerebral ischemia decreased the serum level of IL-10, which was prevented by hUCB-MSC transplantation. The expression of IL-1β, IL-6, IL-10 and TNF-α in the peri-ischemic brain tissues showed similar changes as in the serum. hUCB-MSC transplantation markedly suppressed the infiltration of inflammatory cells, and increased the neuronal density around the ischemic region. Furthermore, hUCB-MSC transplantation significantly decreased the percentage of apoptosis around the ischemic region. CONCLUSION hUCB-MSCs transplantation suppresses inflammatory responses and neuronal apoptosis during the early stage focal cerebral ischemia in rabbits.
Collapse
|
81
|
Abstract
This article introduces the basic concepts of modeling neonatal brain injury and provides background information regarding each of the commonly used types of stem cells. It summarizes the findings of preclinical research testing the therapeutic potential of stem cells in animal models of neonatal brain injury, reports briefly on the status of clinical trials, and discusses the important ongoing issues that need to be addressed before stem cell therapy is used to repair the injured brain.
Collapse
|
82
|
Liu YX, Guo XM, Li JF, Meng Y, Zhang HT, Liu AJ, Li SC, Liu YL, Zhu H, Xue JH, Zhang Y, Zhang ZW. Restoration of tissue damage, and never activity after hypoxia–ischemia by implantation of peripheral blood mononuclear cells. Brain Res 2014; 1546:34-45. [PMID: 24373803 DOI: 10.1016/j.brainres.2013.11.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 10/30/2013] [Accepted: 11/26/2013] [Indexed: 11/25/2022]
|
83
|
Effects of intravenous administration of umbilical cord blood CD34(+) cells in a mouse model of neonatal stroke. Neuroscience 2014; 263:148-58. [PMID: 24444827 DOI: 10.1016/j.neuroscience.2014.01.018] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 01/06/2014] [Accepted: 01/06/2014] [Indexed: 12/16/2022]
Abstract
Neonatal stroke occurs in approximately 1/4000 live births and results in life-long neurological impairments: e.g., cerebral palsy. Currently, there is no evidence-based specific treatment for neonates with stroke. Several studies have reported the benefits of umbilical cord blood (UCB) cell treatment in rodent models of neonatal brain injury. However, all of the studies examined the effects of administering either the UCB mononuclear cell fraction or UCB-derived mesenchymal stem cells in neonatal rat models. The objective of this study was to examine the effects of human UCB CD34(+) cells (hematopoietic stem cell/endothelial progenitor cells) in a mouse model of neonatal stroke, which we recently developed. On postnatal day 12, immunocompromized (SCID) mice underwent permanent occlusion of the left middle cerebral artery (MCAO). Forty-eight hours after MCAO, human UCB CD34(+) cells (1×10(5)cells) were injected intravenously into the mice. The area in which cerebral blood flow (CBF) was maintained was temporarily larger in the cell-treated group than in the phosphate-buffered saline (PBS)-treated group at 24h after treatment. With cell treatment, the percent loss of ipsilateral hemispheric volume was significantly ameliorated (21.5±1.9%) compared with the PBS group (25.6±5.1%) when assessed at 7weeks after MCAO. The cell-treated group did not exhibit significant differences from the PBS group in either rotarod (238±46s in the sham-surgery group, 175±49s in the PBS group, 203±54s in the cell-treated group) or open-field tests. The intravenous administration of human UCB CD34(+) cells modestly reduced histological ischemic brain damage after neonatal stroke in mice, with a transient augmentation of CBF in the peri-infarct area.
Collapse
|
84
|
Abstract
Fetal or neonatal brain injury can result in lifelong neurologic disability. The most significant risk factor for perinatal brain injury is prematurity; however, in absolute numbers, full-term infants represent the majority of affected children. Research on strategies to prevent or mitigate the impact of perinatal brain injury ("perinatal neuroprotection") has established the mitigating roles of magnesium sulfate administration for preterm infants and therapeutic hypothermia for term infants with suspected perinatal brain injury. Banked umbilical cord blood, erythropoietin, and a number of other agents that may improve neuronal repair show promise for improving outcomes following perinatal brain injury in animal models. Other preventative strategies include delayed umbilical cord clamping in preterm infants and progesterone in women with prior preterm birth or short cervix and avoidance of infections. Despite these advances, we have not successfully decreased the rate of preterm birth, nor are we able to predict term infants at risk of hypoxic brain injury in order to intervene prior to the hypoxic event. Further, we lack the ability to modulate the sequelae of neuronal cell insults or the ability to repair brain injury after it has been sustained. As a consequence, despite exciting advances in the field of perinatal neuroprotection, perinatal brain injury still impacts thousands of newborns each year with significant long-term morbidity and mortality.
Collapse
Affiliation(s)
- Kirsten E. Salmeen
- 513 Parnassus Avenue, Room HSE-1634, Box 0556, San Francisco, CA 94143-0556USA
| | - Angie C. Jelin
- 106 Irving Street, NW, Room POB 108, Washington, DC 20010USA
| | - Mari-Paule Thiet
- 505 Parnassus Avenue, Moffitt 1478, Box 0132, San Francisco, CA 94143-0132USA
| |
Collapse
|
85
|
Ruff CA, Faulkner SD, Fehlings MG. The potential for stem cell therapies to have an impact on cerebral palsy: opportunities and limitations. Dev Med Child Neurol 2013; 55:689-97. [PMID: 23680015 DOI: 10.1111/dmcn.12166] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/02/2013] [Indexed: 12/31/2022]
Abstract
Cerebral palsy (CP) is a chronic childhood disorder described by a group of motor and cognitive impairments and results in a substantial socio-economic burden to the individual, family, and healthcare system. With no effective biological interventions, therapies for CP are currently restricted to supportive and management strategies. Cellular transplantation has been suggested as a putative intervention for neural pathology, as mesenchymal and neural stem cells, as well as olfactory ensheathing glia and Schwann cells, have shown some regenerative and functional efficacy in experimental central nervous system disorders. This review describes the most common cell types investigated and delineates their purported mechanisms in vivo. Furthermore, it provides a cogent summary of both current early-phase clinical trials using neural precursor cells (NPCs) and the state of stem cell therapies for neurodegenerative conditions. Although NPCs are perhaps the most promising candidates for cell replacement therapy in the context of CP, much still remains to be understood regarding safety, efficacy, timing, dose, and route of transplantation, as well as the capacity for combinatorial strategies.
Collapse
Affiliation(s)
- Crystal A Ruff
- Division of Genetics and Development, Toronto Western Research Institute, Toronto, ON, Canada
| | | | | |
Collapse
|
86
|
First autologous cell therapy of cerebral palsy caused by hypoxic-ischemic brain damage in a child after cardiac arrest-individual treatment with cord blood. Case Rep Transplant 2013; 2013:951827. [PMID: 23762741 PMCID: PMC3671311 DOI: 10.1155/2013/951827] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 04/18/2013] [Indexed: 01/08/2023] Open
Abstract
Each year, thousands of children incur brain damage that results in lifelong sequelae. Therefore, based on experimental evidence, we explored the therapeutic potential of human cord blood, known to contain stem cells, to examine the functional neuroregeneration in a child with cerebral palsy after cardiac arrest. The boy, whose cord blood was stored at birth, was 2.5 years old and normally developed when global ischemic brain damage occurred resulting in a persistent vegetative state. Nine weeks later, he received autologous cord blood (91.7 mL, cryopreserved, 5.75 × 10e8 mononuclear cells) intravenously. Active rehabilitation (physio- and ergotherapy) was provided daily, follow-up at 2, 5, 12, 24, 30, and 40 months. At 2-months follow-up the boy's motor control improved, spastic paresis was largely reduced, and eyesight was recovered, as did the electroencephalogram. He smiled when played with, was able to sit and to speak simple words. At 40 months, independent eating, walking in gait trainer, crawling, and moving from prone position to free sitting were possible, and there was significantly improved receptive and expressive speech competence (four-word sentences, 200 words). This remarkable functional neuroregeneration is difficult to explain by intense active rehabilitation alone and suggests that autologous cord blood transplantation may be an additional and causative treatment of pediatric cerebral palsy after brain damage.
Collapse
|
87
|
Pluripotent possibilities: human umbilical cord blood cell treatment after neonatal brain injury. Pediatr Neurol 2013; 48:346-54. [PMID: 23583051 DOI: 10.1016/j.pediatrneurol.2012.10.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 10/29/2012] [Indexed: 12/14/2022]
Abstract
Perinatal hypoxic-ischemic brain injury and stroke in the developing brain remain important causes of chronic neurologic morbidity. Emerging data suggest that transplantation of umbilical cord blood-derived stem cells may have therapeutic potential for neuroregeneration and improved functional outcome. The pluripotent capacity of stem cells from the human umbilical cord blood provides simultaneous targeting of multiple neuropathologic events initiated by a hypoxic-ischemic insult. Their high regenerative potential and naïve immunologic phenotype makes them a preferable choice for transplantation. A multiplicity of transplantation protocols have been studied with a variety of brain injury models; however, only a few have been conducted on immature animals. Biological recipient characteristics, such as age and sex, appear to differentially modulate responses of the animals to the transplanted cord blood stem cells. Survival, migration, and function of the transplanted cells have also been studied and reveal insights into the mechanisms of cord blood stem cell effects. Data from preclinical studies have informed current clinical safety trials of human cord blood in neonates, and further work is needed to continue progress in this field.
Collapse
|
88
|
The endogenous regenerative capacity of the damaged newborn brain: boosting neurogenesis with mesenchymal stem cell treatment. J Cereb Blood Flow Metab 2013; 33:625-34. [PMID: 23403379 PMCID: PMC3652688 DOI: 10.1038/jcbfm.2013.3] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Neurogenesis continues throughout adulthood. The neurogenic capacity of the brain increases after injury by, e.g., hypoxia-ischemia. However, it is well known that in many cases brain damage does not resolve spontaneously, indicating that the endogenous regenerative capacity of the brain is insufficient. Neonatal encephalopathy leads to high mortality rates and long-term neurologic deficits in babies worldwide. Therefore, there is an urgent need to develop more efficient therapeutic strategies. The latest findings indicate that stem cells represent a novel therapeutic possibility to improve outcome in models of neonatal encephalopathy. Transplanted stem cells secrete factors that stimulate and maintain neurogenesis, thereby increasing cell proliferation, neuronal differentiation, and functional integration. Understanding the molecular and cellular mechanisms underlying neurogenesis after an insult is crucial for developing tools to enhance the neurogenic capacity of the brain. The aim of this review is to discuss the endogenous capacity of the neonatal brain to regenerate after a cerebral ischemic insult. We present an overview of the molecular and cellular mechanisms underlying endogenous regenerative processes during development as well as after a cerebral ischemic insult. Furthermore, we will consider the potential to use stem cell transplantation as a means to boost endogenous neurogenesis and restore brain function.
Collapse
|
89
|
Wu CC, Chen YC, Chang YC, Wang LW, Lin YC, Chiang YL, Ho CJ, Huang CC. Human umbilical vein endothelial cells protect against hypoxic-ischemic damage in neonatal brain via stromal cell-derived factor 1/C-X-C chemokine receptor type 4. Stroke 2013; 44:1402-9. [PMID: 23449265 DOI: 10.1161/strokeaha.111.000719] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND PURPOSE Agents that protect against neurovascular damage provide a powerful neuroprotective strategy. Human umbilical vein endothelial cells (HUVECs) may be used to treat neonates with hypoxic-ischemia (HI) because of its autologous capability. We hypothesized that peripherally injected HUVECs entered the brain after HI, protected against neurovascular damage, and provided protection via stromal cell-derived factor 1/C-X-C chemokine receptor type 4 pathway in neonatal brain. METHODS Postpartum day 7 rat pups received intraperitoneal injections of low-passage HUVEC-P4, high-passage HUVEC-P8, or conditioned medium before and immediately after HI. HUVECs were transfected with adenovirus-green fluorescent protein for cell tracing. Oxygen-glucose deprivation was established by coculturing HUVEC-P4 with mouse neuroblastoma neuronal cells (Neuro-2a) and with mouse immortalized cerebral vascular endothelial cells (b.End3). RESULTS HUVEC-P4-treated group had more blood levels of green fluorescent protein-positive cells than HUVEC-P8-treated group 3 hours postinjection. Intraperitoneally injected HUVEC-P4, but not HUVEC-P8, entered the cortex after HI and positioned closed to the neurons and microvessels. Compared with the condition medium-treated group, the HUVEC-P4-treated but not the HUVEC-P8-treated group showed significantly less neuronal apoptosis and blood-brain barrier damage and more preservation of microvessels in the cortex 24 hours after HI. On postpartum day 14, the HUVEC-P4-treated group showed significant neuroprotection compared with the condition medium-treated group. Stromal cell-derived factor 1 was upregulated in the ipsilateral cortex 3 hours after HI, and inhibiting the stromal cell-derived factor 1/C-X-C chemokine receptor type 4 reduced the protective effect of HUVEC-P4. In vitro transwell coculturing of HUVEC-P4 also significantly protected against oxygen-glucose deprivation cell death in neurons and endothelial cells. CONCLUSIONS Cell therapy using HUVECs may provide a powerful therapeutic strategy in treating neonates with HI.
Collapse
Affiliation(s)
- Chia-Ching Wu
- Department of Cell Biology and Anatomy, National Cheng Kung University Hospital, No. 138 Sheng-Li Rd, Tainan City 704, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Dalous J, Pansiot J, Pham H, Chatel P, Nadaradja C, D'Agostino I, Vottier G, Schwendimann L, Vanneaux V, Charriaut-Marlangue C, Titomanlio L, Gressens P, Larghero J, Baud O. Use of Human Umbilical Cord Blood Mononuclear Cells to Prevent Perinatal Brain Injury: A Preclinical Study. Stem Cells Dev 2013; 22:169-79. [DOI: 10.1089/scd.2012.0183] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Jérémie Dalous
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Julien Pansiot
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Hoa Pham
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Paul Chatel
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Céline Nadaradja
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Irene D'Agostino
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Gaëlle Vottier
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Leslie Schwendimann
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Valérie Vanneaux
- Unité de Thérapie Cellulaire et Centre d'Investigation Clinique en Biothérapies CIC-BT501, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Christiane Charriaut-Marlangue
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Luigi Titomanlio
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Pierre Gressens
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Jérôme Larghero
- Unité de Thérapie Cellulaire et Centre d'Investigation Clinique en Biothérapies CIC-BT501, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Olivier Baud
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
- NICU, Université Paris-Diderot, Hôpital Robert Debré, APHP, Paris, France
| |
Collapse
|
91
|
Repairing neural injuries using human umbilical cord blood. Mol Neurobiol 2012; 47:938-45. [PMID: 23275174 DOI: 10.1007/s12035-012-8388-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 12/13/2012] [Indexed: 01/14/2023]
Abstract
Stem cells are promising sources for repairing damaged neurons and glial cells in neural injuries and for replacing dead cells in neurodegenerative diseases. An essential step for stem cell-based therapy is to generate large quantities of stem cells and develop reliable culture conditions to direct efficient differentiation of specific neuronal and glial subtypes. The human umbilical cord and umbilical cord blood (UCB) are rich sources of multiple stem cells, including hematopoietic stem cells, mesenchymal stem cells, unrestricted somatic stem cells, and embryonic-like stem cells. Human UC/UCB-derived cells are able to give rise to multiple cell types of neural lineages. Studies have shown that UCB and UCB-derived cells can survive in injured sites in animal models of ischemic brain damage and spinal cord injuries, and promote survival and prevent cell death of local neurons and glia. Human UCB is easy to harvest and purify. Moreover, unlike embryonic stem cells, the use of human UCB is not limited by ethical quandaries. Therefore, human UCB is an attractive source of stem cells for repairing neural injuries.
Collapse
|
92
|
Rosenkranz K, May C, Meier C, Marcus K. Proteomic analysis of alterations induced by perinatal hypoxic-ischemic brain injury. J Proteome Res 2012; 11:5794-803. [PMID: 23153068 DOI: 10.1021/pr3005869] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Perinatal hypoxic-ischemic brain injury is an important cause of neurological deficits still causing mortality and morbidity in the early period of life. As efficient clinical or pharmaceutical strategies to prevent or reduce the outcome of perinatal hypoxic-ischemic brain damage are limited, the development of new therapies is of utmost importance. To evolve innovative therapeutic concepts, elucidation of the mechanisms contributing to the neurological impairments upon hypoxic-ischemic brain injury is necessary. Therefore, we aimed for the identification of proteins that are affected by hypoxic-ischemic brain injury in neonatal rats. To assess changes in protein expression two days after induction of brain damage, a 2D-DIGE based proteome analysis was performed. Among the proteins altered after hypoxic-ischemic brain injury, Calcineurin A, Coronin-1A, as well as GFAP were identified, showing higher expression in lesioned hemispheres. Validation of the changes in Calcineurin A expression by Western Blot analysis demonstrated several truncated forms of this protein generated by limited proteolysis after hypoxia-ischemia. Further analysis revealed activation of calpain, which is involved in the limited proteolysis of Calcineurin. Active forms of Calcineurin are associated with the dephosphorylation of Darpp-32, an effect that was also demonstrated in lesioned hemispheres after perinatal brain injury.
Collapse
Affiliation(s)
- Katja Rosenkranz
- Department of Functional Proteomics, Ruhr-University Bochum, Germany.
| | | | | | | |
Collapse
|
93
|
Rosenkranz K, Tenbusch M, May C, Marcus K, Meier C. Changes in Interleukin-1 alpha serum levels after transplantation of umbilical cord blood cells in a model of perinatal hypoxic-ischemic brain damage. Ann Anat 2012; 195:122-7. [PMID: 23123184 DOI: 10.1016/j.aanat.2012.09.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 08/21/2012] [Accepted: 09/15/2012] [Indexed: 01/07/2023]
Abstract
Transplantation of human umbilical cord blood (hUCB) cells is a potential approach for the treatment of perinatal hypoxic-ischemic brain injury. Neurological and motor deficits resulting from the brain lesion are ameliorated upon transplantation. The molecular mechanisms underlying these improvements are currently being unravelled. One parameter identified as part of the beneficial effects of hUCB cells is the reduction of brain inflammation. It is, however, unclear whether the modulation of brain inflammation is due to local or systemic effects of hUCB cells. In this study, the effects of hUCB cell transplantation in a model of perinatal hypoxic-ischemic brain injury were investigated at the systemic level by measurement of serum levels of pro-inflammatory cytokines by multiplex bead arrays. Two days after induction of the brain damage, levels of the pro-inflammatory cytokines Interleukin-1α (IL-1α), Interleukin-1β (IL-1β), and Tumor necrosis factor α (TNFα) were increased in the serum of rats. Application of hUCB cells, in turn, correlated with a reduced elevation of serum levels of these pro-inflammatory cytokines. This decrease was accompanied by a reduced expression of CD68, a marker protein of activated microglia/macrophages in the brain. Therefore, systemic modulation of the immune response by hUCB cells could represent one possible mechanism of how these cells might mediate their beneficial effects. Creation of a regenerative environment with reduced inflammation might account for the functional regeneration observed upon hUCB cell treatment in lesioned animals.
Collapse
Affiliation(s)
- Katja Rosenkranz
- Department of Functional Proteomics, Ruhr-University Bochum, Germany.
| | | | | | | | | |
Collapse
|
94
|
Rescuing the neonatal brain from hypoxic injury with autologous cord blood. Bone Marrow Transplant 2012; 48:890-900. [PMID: 22964590 DOI: 10.1038/bmt.2012.169] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 08/02/2012] [Accepted: 08/03/2012] [Indexed: 12/28/2022]
Abstract
Brain injury resulting from perinatal hypoxic-ischemic encephalopathy (HIE) is a major cause of acute mortality in infants and chronic neurologic disability in surviving children. Recent multicenter clinical trials demonstrated the effectiveness of hypothermia initiated within the first 6 postnatal hours to reduce the risk of death or major neurological disabilities among neonates with HIE. However, in these trials, approximately 40% of cooled infants died or survived with significant impairments. Therefore, adjunct therapies are required to improve the outcome in neonates with HIE. Cord blood (CB) is a rich source of stem cells. Administration of human CB cells in animal models of HIE has generally resulted in improved outcomes and multiple mechanisms have been suggested including anti-inflammation, release of neurotrophic factors and stimulation of endogenous neurogenesis. Investigators at Duke are conducting studies of autologous CB infusion in neonates with HIE and in children with cerebral palsy. These pilot studies indicate no added risk from the regimens used, but results of ongoing placebo-controlled trials are needed to assess efficacy. Meanwhile, further investigations are warranted to determine the best strategies, that is, timing, dosing, route of delivery, choice of stem cells and ex vivo modulations, to attain long-term benefits of CB stem cell therapy.
Collapse
|
95
|
Moraes L, Vasconcelos-dos-Santos A, Santana FC, Godoy MA, Rosado-de-Castro PH, Jasmin, Azevedo-Pereira RL, Cintra WM, Gasparetto EL, Santiago MF, Mendez-Otero R. Neuroprotective effects and magnetic resonance imaging of mesenchymal stem cells labeled with SPION in a rat model of Huntington's disease. Stem Cell Res 2012; 9:143-55. [DOI: 10.1016/j.scr.2012.05.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 04/26/2012] [Accepted: 05/18/2012] [Indexed: 01/14/2023] Open
|
96
|
Wasielewski B, Jensen A, Roth-Härer A, Dermietzel R, Meier C. Neuroglial activation and Cx43 expression are reduced upon transplantation of human umbilical cord blood cells after perinatal hypoxic-ischemic injury. Brain Res 2012; 1487:39-53. [PMID: 22796290 DOI: 10.1016/j.brainres.2012.05.066] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2012] [Accepted: 05/31/2012] [Indexed: 12/15/2022]
Abstract
Glial cells play a crucial role in the pathomechanism of perinatal hypoxic-ischemic brain injury (HI) and are involved in the maintenance of a chronic state of inflammation that causes delayed neuronal damage. Activation of astrocytes is one factor prolonging brain damage and contributing to the formation of a glial scar that limits neuronal plasticity. In this context, the major astrocytic gap junction protein Connexin 43 (Cx43) has been ascribed various functions including regulation of astrocytic migration and proliferation. Here, we investigate glial responses like microglia/macrophages and astrocytic activation in a rat model of neonatal HI and characterize changes of these parameters upon transplantation of human umbilical cord blood cells (hUCB). As an alleviation of motor function in lesioned rats has previously been described in transplanted animals, we analyze the putative correlation between motor function and glial activation over time. The lesion-induced impairment of motor function, assessed by forelimb use bias, muscle strength and distal spasticity, was alleviated upon transplantation of hUCB short and long term. HI induced an acute inflammatory reaction with activation of microglia/macrophages and reactive astrogliosis associated with perilesional upregulation of Cx43 that slowly declined during the chronic post-ischemic phase. hUCB transplantation accelerated the regression of inflammatory events, narrowed the perilesional astrocytic wall and led to a downregulation of the investigated astrocytic proteins. Thus, in the immature brain, hUCB may indirectly reduce secondary cell death upon hypoxia-ischemia and facilitate post-ischemic plasticity through the attenuation of reactive gliosis. This article is part of a Special Issue entitled Electrical Synapses.
Collapse
Affiliation(s)
- Bianca Wasielewski
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, D-44801 Bochum, Germany
| | | | | | | | | |
Collapse
|
97
|
Bennet L, Tan S, Van den Heuij L, Derrick M, Groenendaal F, van Bel F, Juul S, Back SA, Northington F, Robertson NJ, Mallard C, Gunn AJ. Cell therapy for neonatal hypoxia-ischemia and cerebral palsy. Ann Neurol 2012; 71:589-600. [PMID: 22522476 DOI: 10.1002/ana.22670] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Perinatal hypoxic-ischemic brain injury remains a major cause of cerebral palsy. Although therapeutic hypothermia is now established to improve recovery from hypoxia-ischemia (HI) at term, many infants continue to survive with disability, and hypothermia has not yet been tested in preterm infants. There is increasing evidence from in vitro and in vivo preclinical studies that stem/progenitor cells may have multiple beneficial effects on outcome after hypoxic-ischemic injury. Stem/progenitor cells have shown great promise in animal studies in decreasing neurological impairment; however, the mechanisms of action of stem cells, and the optimal type, dose, and method of administration remain surprisingly unclear, and some studies have found no benefit. Although cell-based interventions after completion of the majority of secondary cell death appear to have potential to improve functional outcome for neonates after HI, further rigorous testing in translational animal models is required before randomized controlled trials should be considered.
Collapse
Affiliation(s)
- Laura Bennet
- Department of Physiology, University of Auckland, New Zealand
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Intravenous administration of human umbilical cord blood-mononuclear cells dose-dependently relieve neurologic deficits in rat intracerebral hemorrhage model. Ann Anat 2012; 195:39-49. [PMID: 22770555 DOI: 10.1016/j.aanat.2012.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 05/05/2012] [Accepted: 05/07/2012] [Indexed: 01/01/2023]
Abstract
Human umbilical cord blood (HUCB) is now considered as a valuable source for stem cell-based therapies. Previous studies showed that intravascular injection of the HUCB significantly improves neurological functional recovery in a model of intracerebral hemorrhage (ICH). To extend these findings, we examined the behavioral recovery and injured volume in the presence of increasing doses of human umbilical cord blood derived mononuclear cells (HUC-MCs) after intracerebral hemorrhage in rats. The experimental ICH was induced by intrastriatal administration of bacterial collagenase IV in adult rats. One day after the surgery, the rats were randomly divided into 4 groups to receive intravenously either BrdU positive human UC-MCs (4 × 10(6), 8 × 10(6) and 16 × 10(6) cells in 1 ml saline, n=10, respectively) as treated groups or the same amount of saline as lesion group (n=10). There was also one group (control n=10) that received only the vehicle solution of collagenase. The animals were evaluated for 14 days with modified limb placing and corner turn tests. The transplanted human UC-MCs were also detected by immunohistochemistry with labeling of BrdU. Two weeks after infusion, there was a significant recovery in the behavioral performance when 4 × 10(6) or more UC-MCs were delivered (P<0.05-0.001). Injured volume measurements disclosed an inverse relationship between UC-MCs dose and damage reaching significance at the higher UC-MCs doses. Moreover, human UC-MCs were localized by immunohistochemistry only in the injured area. Intravenously transplanted UC-MCs can accelerate the neurological function recovery of ICH rat and diminish the striatum lesion size by demonstrating a dose relationship between them.
Collapse
|
99
|
Rosenkranz K, Kumbruch S, Tenbusch M, Marcus K, Marschner K, Dermietzel R, Meier C. Transplantation of human umbilical cord blood cells mediated beneficial effects on apoptosis, angiogenesis and neuronal survival after hypoxic-ischemic brain injury in rats. Cell Tissue Res 2012; 348:429-38. [PMID: 22526623 DOI: 10.1007/s00441-012-1401-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 03/07/2012] [Indexed: 12/31/2022]
Abstract
Transplantation of human umbilical cord blood (hucb) cells in a model of hypoxic-ischemic brain injury led to the amelioration of lesion-impaired neurological and motor functions. However, the mechanisms by which transplanted cells mediate functional recovery after brain injury are largely unknown. In this study, the effects of hucb cell transplantation were investigated in this experimental paradigm at the cellular and molecular level. As the pathological cascade in hypoxic-ischemic brain injury includes inflammation, reduced blood flow, and neuronal cell death, we analyzed the effects of peripherally administered hucb cells on these detrimental processes, investigating the expression of characteristic marker proteins. Application of hucb cells after perinatal hypoxic-ischemic brain injury correlated with an increased expression of the proteins Tie-2 and occludin, which are associated with angiogenesis. Lesion-induced apoptosis, determined by expression of cleaved caspase-3, decreased, whereas the number of vital neurons, identified by counting of NeuN-positive cells, increased. In addition, we observed an increase in the expression of neurotrophic and pro-angiogenic growth factors, namely BDNF and VEGF, in the lesioned brain upon hucb cell transplantation. The release of neurotrophic factors mediated by transplanted hucb cells might cause a lower number of neurons to undergo apoptosis and result in a higher number of living neurons. In parallel, the increase of VEGF might cause growth of blood vessels. Thus, hucb transplantation might contribute to functional recovery after brain injury mediated by systemic or local effects.
Collapse
Affiliation(s)
- Katja Rosenkranz
- Department of Functional Proteomics, Ruhr-University Bochum, Bochum, Germany
| | | | | | | | | | | | | |
Collapse
|
100
|
Bae SH, Kong TH, Lee HS, Kim KS, Hong KS, Chopp M, Kang MS, Moon J. Long-lasting paracrine effects of human cord blood cells on damaged neocortex in an animal model of cerebral palsy. Cell Transplant 2012; 21:2497-515. [PMID: 22524897 DOI: 10.3727/096368912x640457] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Neonatal asphyxia is an important contributor to cerebral palsy (CP), for which there is no effective treatment to date. The administration of human cord blood cells (hUCBCs) is emerging as a therapeutic strategy for the treatment of neurological disorders. However, there are few studies on the application of hUCBCs to the treatment of neonatal ischemia as a model of CP. Experiments and behavioral tests (mainly motor tests) performed on neonatal hypoxia/ischemia have been limited to short-term effects of hUCBCs, but mechanisms of action have not been investigated. We performed a study on the use of hUCBCs in a rat model of neonatal hypoxia/ischemia and investigated the underlying mechanism for therapeutic benefits of hUCBC treatment. hUCBCs were intravenously transplanted into a rat model of neonatal hypoxia ischemia. hUCBCs increased microglia temporarily in the periventricular striatum in the early phase of disease, protected mature neurons in the neocortex from injury, paved the way for the near-normalization of brain damage in the subventricular zone (SVZ), and, in consequence, significantly improved performance in a battery of behavioral tests compared to the vehicle-treated group. Although the transplanted cells were rarely observed in the brain 3 weeks after transplantation, the effects of the improved behavioral functions persisted. Our preclinical findings suggest that the long-lasting positive influence of hUCBCs is derived from paracrine effects of hUCBCs that stimulate recovery in the injured brain and protect against further brain damage.
Collapse
Affiliation(s)
- Sang-Hun Bae
- College of Life Science, Department of Applied Bioscience, CHA University, Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|