Mesenchymal Stem Cells Induce T-Cell Tolerance and Protect the Preterm Brain after Global Hypoxia-Ischemia

Hypoxic-ischemic encephalopathy (HIE) in preterm infants is a severe disease for which no curative treatment is available. Cerebral inflammation and invasion of activated peripheral immune cells have been shown to play a pivotal role in the etiology of white matter injury, which is the clinical hallmark of HIE in preterm infants. The objective of this study was to assess the neuroprotective and anti-inflammatory effects of intravenously delivered mesenchymal stem cells (MSC) in an ovine model of HIE. In this translational animal model, global hypoxia-ischemia (HI) was induced in instrumented preterm sheep by transient umbilical cord occlusion, which closely mimics the clinical insult. Intravenous administration of 2 x 106 MSC/kg reduced microglial proliferation, diminished loss of oligodendrocytes and reduced demyelination, as determined by histology and Diffusion Tensor Imaging (DTI), in the preterm brain after global HI. These anti-inflammatory and neuroprotective effects of MSC were paralleled by reduced electrographic seizure activity in the ischemic preterm brain. Furthermore, we showed that MSC induced persistent peripheral T-cell tolerance in vivo and reduced invasion of T-cells into the preterm brain following global HI. These findings show in a preclinical animal model that intravenously administered MSC reduced cerebral inflammation, protected against white matter injury and established functional improvement in the preterm brain following global HI. Moreover, we provide evidence that induction of T-cell tolerance by MSC might play an important role in the neuroprotective effects of MSC in HIE. This is the first study to describe a marked neuroprotective effect of MSC in a translational animal model of HIE.

[1]  D B Vigneron,et al.  Seizure-associated brain injury in term newborns with perinatal asphyxia , 2002, Neurology.

[2]  Alan W. Flake,et al.  Human mesenchymal stem cells engraft and demonstrate site-specific differentiation after in utero transplantation in sheep , 2000, Nature Medicine.

[3]  M. Kas,et al.  Intranasal Mesenchymal Stem Cell Treatment for Neonatal Brain Damage: Long-Term Cognitive and Sensorimotor Improvement , 2013, PloS one.

[4]  Peter Andriessen,et al.  Cerebral inflammation and mobilization of the peripheral immune system following global hypoxia-ischemia in preterm sheep , 2013, Journal of Neuroinflammation.

[5]  M. Pallavicini,et al.  Engraftment and long-term expression of human fetal hemopoietic stem cells in sheep following transplantation in utero. , 1992, The Journal of clinical investigation.

[6]  J. Volpe Neurology of the Newborn , 1959, Major problems in clinical pediatrics.

[7]  L. D. de Vries,et al.  Antemortem cranial MRI compared with postmortem histopathologic examination of the brain in term infants with neonatal encephalopathy following perinatal asphyxia , 2012, Archives of Disease in Childhood: Fetal and Neonatal Edition.

[8]  Reint K Jellema,et al.  Inflammation-induced immune suppression of the fetus: a potential link between chorioamnionitis and postnatal early onset sepsis , 2012, The journal of maternal-fetal & neonatal medicine : the official journal of the European Association of Perinatal Medicine, the Federation of Asia and Oceania Perinatal Societies, the International Society of Perinatal Obstetricians.

[9]  Chang Won Choi,et al.  Mesenchymal Stem-Cell Transplantation for Hypoxic-Ischemic Brain Injury in Neonatal Rat Model , 2010, Pediatric Research.

[10]  A. Uccelli,et al.  Systemic Administration of Mesenchymal Stem Cells Increases Neuron Survival after Global Cerebral Ischemia In Vivo (2VO) , 2010, Neural plasticity.

[11]  M. Pittenger,et al.  Human mesenchymal stem cells modulate allogeneic immune cell responses. , 2005, Blood.

[12]  H. Kinney,et al.  Late Oligodendrocyte Progenitors Coincide with the Developmental Window of Vulnerability for Human Perinatal White Matter Injury , 2001, The Journal of Neuroscience.

[13]  Jean P O'Malley,et al.  Selective vulnerability of preterm white matter to oxidative damage defined by F2‐isoprostanes , 2005, Annals of neurology.

[14]  A. Gunn,et al.  Cell therapy for neonatal hypoxia–ischemia and cerebral palsy , 2012, Annals of neurology.

[15]  Oh Young Bang,et al.  Autologous mesenchymal stem cell transplantation in stroke patients , 2005, Annals of neurology.

[16]  Steven P. Miller,et al.  Clinical Neonatal Seizures are Independently Associated with Outcome in Infants at Risk for Hypoxic-Ischemic Brain Injury. , 2009, The Journal of pediatrics.

[17]  Justin Dean,et al.  Histopathological correlates of magnetic resonance imaging–defined chronic perinatal white matter injury , 2011, Annals of neurology.

[18]  N. Rouas-Freiss,et al.  Human Leukocyte Antigen‐G5 Secretion by Human Mesenchymal Stem Cells Is Required to Suppress T Lymphocyte and Natural Killer Function and to Induce CD4+CD25highFOXP3+ Regulatory T Cells , 2008, Stem cells.

[19]  A. Gunn,et al.  The effect of cerebral hypothermia on white and grey matter injury induced by severe hypoxia in preterm fetal sheep , 2007, The Journal of physiology.

[20]  C. Thiemermann,et al.  Mesenchymal Stromal Cells: Current Understanding and Clinical Status , 2009, Stem cells.

[21]  Nathan J Stevenson,et al.  The temporal evolution of electrographic seizure burden in neonatal hypoxic ischemic encephalopathy , 2012, Epilepsia.

[22]  A. Gunn,et al.  Relationship between evolving epileptiform activity and delayed loss of mitochondrial activity after asphyxia measured by near‐infrared spectroscopy in preterm fetal sheep , 2006, The Journal of physiology.

[23]  L. Aigner,et al.  Mesenchymal stem cells prime proliferating adult neural progenitors toward an oligodendrocyte fate. , 2012, Stem cells and development.

[24]  D. Hess,et al.  Intravenous Grafts Recapitulate the Neurorestoration Afforded by Intracerebrally Delivered Multipotent Adult Progenitor Cells in Neonatal Hypoxic-Ischemic Rats , 2008, Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism.

[25]  A. Kavelaars,et al.  Mesenchymal stem cell transplantation changes the gene expression profile of the neonatal ischemic brain , 2011, Brain, Behavior, and Immunity.

[26]  P. Delafontaine,et al.  Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. , 2009, Cell stem cell.

[27]  J. Volpe Brain injury in premature infants: a complex amalgam of destructive and developmental disturbances , 2009, The Lancet Neurology.

[28]  Yuji Shen,et al.  Reduced Fractional Anisotropy on Diffusion Tensor Magnetic Resonance Imaging After Hypoxic-Ischemic Encephalopathy , 2006, Pediatrics.

[29]  P. Hurn,et al.  Experimental Stroke Induces Massive, Rapid Activation of the Peripheral Immune System , 2006, Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism.

[30]  Andrew Whitelaw,et al.  Selective head cooling with mild systemic hypothermia after neonatal encephalopathy: multicentre randomised trial , 2005, The Lancet.

[31]  Peter Andriessen,et al.  Multi-channel amplitude-integrated EEG characteristics in preterm infants with a normal neurodevelopment at two years of corrected age. , 2012, Early human development.

[32]  C. Lois,et al.  Mesenchymal Stem Cells Instruct Oligodendrogenic Fate Decision on Adult Neural Stem Cells , 2006, Stem cells.

[33]  K. Pennypacker,et al.  The spleen contributes to stroke‐induced neurodegeneration , 2008, Journal of neuroscience research.

[34]  D. Hess,et al.  Behavioral and Histological Characterization of Intrahippocampal Grafts of Human Bone Marrow-Derived Multipotent Progenitor Cells in Neonatal Rats with Hypoxic-Ischemic Injury , 2006, Cell transplantation.

[35]  Reint K Jellema,et al.  Regenerative Therapies in Neonatology: Clinical Perspectives , 2012, Klinische Pädiatrie.

[36]  T. Sobrino,et al.  Inflammatory and Neuroimmunomodulatory Changes in Acute Cerebral Ischemia , 2009, Cerebrovascular Diseases.

[37]  U. Ådén,et al.  Long Lasting Local and Systemic Inflammation after Cerebral Hypoxic ischemia in Newborn Mice , 2012, PloS one.

[38]  Jan Sijbers,et al.  ExploreDTI: a graphical toolbox for processing, analyzing, and visualizing diffusion MR data , 2009 .

[39]  Alistair J. Gunn,et al.  Maternal dexamethasone and EEG hyperactivity in preterm fetal sheep , 2011, The Journal of physiology.

[40]  I. Rosén The physiological basis for continuous electroencephalogram monitoring in the neonate. , 2006, Clinics in perinatology.

[41]  P. Hurn,et al.  Effect of experimental stroke on peripheral immunity: CNS ischemia induces profound immunosuppression , 2009, Neuroscience.

[42]  Harner Rn,et al.  Mechanisms of electroencephalographic and behavioral changes produced by parenteral L-DOPA. , 1969 .

[43]  A. Uccelli,et al.  Mesenchymal stem cells in health and disease , 2008, Nature Reviews Immunology.

[44]  Steven P. Miller,et al.  Arrested preoligodendrocyte maturation contributes to myelination failure in premature infants , 2012, Annals of neurology.

[45]  Frank van Bel,et al.  Mesenchymal stem cells restore cortical rewiring after neonatal ischemia in mice , 2012, Annals of neurology.

[46]  Christian Gerloff,et al.  Temporal and spatial dynamics of cerebral immune cell accumulation in stroke. , 2009, Stroke.

[47]  A. Kavelaars,et al.  Repeated Mesenchymal Stem Cell Treatment after Neonatal Hypoxia–Ischemia Has Distinct Effects on Formation and Maturation of New Neurons and Oligodendrocytes Leading to Restoration of Damage, Corticospinal Motor Tract Activity, and Sensorimotor Function , 2010, The Journal of Neuroscience.

[48]  Frank van Bel,et al.  Mesenchymal stem cell treatment after neonatal hypoxic-ischemic brain injury improves behavioral outcome and induces neuronal and oligodendrocyte regeneration , 2010, Brain, Behavior, and Immunity.

[49]  G. Lenzi,et al.  Imaging of Carotid Plaque Angiogenesis , 2009, Cerebrovascular Diseases.

[50]  C. Carlo-Stella,et al.  Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. , 2002, Blood.

[51]  J. Arends,et al.  Morphologically normal, CD30‐negative B‐lymphocytes with chromosome aberrations in classical Hodgkin's disease: the progenitor cell of the malignant clone? , 1999, The Journal of pathology.

[52]  D. Peebles,et al.  Models of white matter injury: comparison of infectious, hypoxic-ischemic, and excitotoxic insults. , 2002, Mental retardation and developmental disabilities research reviews.

[53]  D. Maynard,et al.  A continuous monitoring device for cerebral activity. , 1969, Electroencephalography and clinical neurophysiology.

[54]  Hannah C. Kinney,et al.  The developing oligodendrocyte: key cellular target in brain injury in the premature infant , 2011, International Journal of Developmental Neuroscience.