CC BY-NC-ND 4.0 · Am J Perinatol 2022; 39(16): 1754-1763
DOI: 10.1055/s-0041-1726451
Review Article

Preventing Brain Damage from Hypoxic–Ischemic Encephalopathy in Neonates: Update on Mesenchymal Stromal Cells and Umbilical Cord Blood Cells

Makoto Nabetani
1   Department of Pediatrics, Yodogawa Christian Hospital, Osaka, Japan
,
2   Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
,
Haruo Shintaku
3   Department of Pediatrics, Faculty of Medicine, Osaka City University, Osaka, Japan
› Author Affiliations
Funding H.S. received public grant from Japan Agency for Medical Research and Development for autologous UCB stem cell therapy for neonatal HIE in Japan.

Abstract

Neonatal hypoxic–ischemic encephalopathy (HIE) causes permanent motor deficit “cerebral palsy (CP),” and may result in significant disability and death. Therapeutic hypothermia (TH) had been established as the first effective therapy for neonates with HIE; however, TH must be initiated within the first 6 hours after birth, and the number needed to treat is from 9 to 11 to prevent brain damage from HIE. Therefore, additional therapies for HIE are highly needed. In this review, we provide an introduction on the mechanisms of HIE cascade and how TH and cell therapies such as umbilical cord blood cells and mesenchymal stromal cells (MSCs), especially umbilical cord-derived MSCs (UC-MSCs), may protect the brain in newborns, and discuss recent progress in regenerative therapies using UC-MSCs for neurological disorders.

The brain damage process “HIE cascade” was divided into six stages: (1) energy depletion, (2) impairment of microglia, (3) inflammation, (4) excitotoxity, (5) oxidative stress, and (6) apoptosis in capillary, glia, synapse and/or neuron. The authors showed recent 13 clinical trials using UC-MSCs for neurological disorders.

The authors suggest that the next step will include reaching a consensus on cell therapies for HIE and establishment of effective protocols for cell therapy for HIE.

Key Points

  • This study includes new insights about cell therapy for neonatal HIE and CP in schema.

  • This study shows precise mechanism of neonatal HIE cascade.

  • The mechanism of cell therapy by comparing umbilical cord blood stem cell with MSC is shown.

  • The review of recent clinical trials of UC-MSC is shown.

Authors' Contributions

M.N. and H.S conceptualized the manuscript; T.M. was involved in methodology and software; T.M. and H.S. were involved in validation. M.N. wrote the original draft and was involved in formal analysis and investigation, and manuscript preparation, review, and editing. H.S. was involved in supervision, project administration, and funding acquisition. All authors have read and agreed to the published version of the manuscript.




Publication History

Received: 05 October 2020

Accepted: 11 February 2021

Article published online:
14 April 2021

© 2021. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/)

Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

 
  • References

  • 1 Jacobs S, Hunt R, Tarnow-Mordi W, Inder T, Davis P. Cooling for newborns with hypoxic ischaemic encephalopathy. Cochrane Database Syst Rev 2007; (04) CD003311
  • 2 Nabetani M, Shintaku H, Hamazaki T. Future perspectives of cell therapy for neonatal hypoxic-ischemic encephalopathy. Pediatr Res 2018; 83 (1-2): 356-363
  • 3 Khoury M, Cuenca J, Cruz FF, Figueroa FE, Rocco PRM, Weiss DJ. Current status of cell-based therapies for respiratory virus infections: applicability to COVID-19. Eur Respir J 2020; 55 (06) 2000858
  • 4 Perlman JM. Intervention strategies for neonatal hypoxic-ischemic cerebral injury. Clin Ther 2006; 28 (09) 1353-1365
  • 5 Volpe JJ. Hypoxic-Ischemic Encephalopathy: Neuropathology and Pathogenesis. Neurology of the Newborn. 5th edition. Philadelphia: Saunders; 2008: 347-399
  • 6 Nabetani M, Okada Y, Kawai S, Nakamura H. Neural activity and the levels of high energy phosphates during deprivation of oxygen and/or glucose in hippocampal slices of immature and adult rats. Int J Dev Neurosci 1995; 13 (01) 3-12
  • 7 Olney JW, Sharpe LG. Brain lesions in an infant rhesus monkey treated with monosodium glutamate. Science 1969; 166 (3903): 386-388
  • 8 Olney JW, Ho OL. Brain damage in infant mice following oral intake of glutamate, aspartate or cysteine. Nature 1970; 227 (5258): 609-611
  • 9 Kirino T. Delayed neuronal death in the gerbil hippocampus following ischemia. Brain Res 1982; 239 (01) 57-69
  • 10 Delpy DT, Gordon RE, Hope PL. et al. Noninvasive investigation of cerebral ischemia by phosphorus nuclear magnetic resonance. Pediatrics 1982; 70 (02) 310-313
  • 11 Hope PL, Costello AM, Cady EB. et al. Cerebral energy metabolism studied with phosphorus NMR spectroscopy in normal and birth-asphyxiated infants. Lancet 1984; 2 (8399): 366-370
  • 12 Blumberg RM, Cady EB, Wigglesworth JS, McKenzie JE, Edwards AD. Relation between delayed impairment of cerebral energy metabolism and infarction following transient focal hypoxia-ischaemia in the developing brain. Exp Brain Res 1997; 113 (01) 130-137
  • 13 Lorek A, Takei Y, Cady EB. et al. Delayed (“secondary”) cerebral energy failure after acute hypoxia-ischemia in the newborn piglet: continuous 48-hour studies by phosphorus magnetic resonance spectroscopy. Pediatr Res 1994; 36 (06) 699-706
  • 14 Simon RP, Swan JH, Griffiths T, Meldrum BS. Blockade of N-methyl-D-aspartate receptors may protect against ischemic damage in the brain. Science 1984; 226 (4676): 850-852
  • 15 Nabetani M, Okada Y, Takata T, Takada S, Nakamura H. Neural activity and intracellular Ca2+ mobilization in the CA1 area of hippocampal slices from immature and mature rats during ischemia or glucose deprivation. Brain Res 1997; 769 (01) 158-162
  • 16 Wada H, Okada Y, Nabetani M, Nakamura H. The effects of lactate and beta-hydroxybutyrate on the energy metabolism and neural activity of hippocampal slices from adult and immature rat. Brain Res Dev Brain Res 1997; 101 (1-2): 1-7
  • 17 Saitoh M, Okada Y, Nabetani M. Effect of mannose, fructose and lactate on the preservation of synaptic potentials in hippocampal slices. Neurosci Lett 1994; 171 (1-2): 125-128
  • 18 Cheng J, Korte N, Nortley R, Sethi H, Tang Y, Attwell D. Targeting pericytes for therapeutic approaches to neurological disorders. Acta Neuropathol 2018; 136 (04) 507-523
  • 19 McCord JM. Oxygen-derived free radicals in postischemic tissue injury. N Engl J Med 1985; 312 (03) 159-163
  • 20 Ferriero DM. Neonatal brain injury. N Engl J Med 2004; 351 (19) 1985-1995
  • 21 Johnston MV, Fatemi A, Wilson MA, Northington F. Treatment advances in neonatal neuroprotection and neurointensive care. Lancet Neurol 2011; 10 (04) 372-382
  • 22 Ikegami A, Haruwaka K, Wake H. Microglia: Lifelong modulator of neural circuits. Neuropathology 2019; 39 (03) 173-180
  • 23 Busto R, Dietrich WD, Globus MY, Ginsberg MD. Postischemic moderate hypothermia inhibits CA1 hippocampal ischemic neuronal injury. Neurosci Lett 1989; 101 (03) 299-304
  • 24 Thoresen M, Bågenholm R, Løberg EM, Apricena F, Kjellmer I. Posthypoxic cooling of neonatal rats provides protection against brain injury. Arch Dis Child Fetal Neonatal Ed 1996; 74 (01) F3-F9
  • 25 Sirimanne ES, Blumberg RM, Bossano D. et al. The effect of prolonged modification of cerebral temperature on outcome after hypoxic-ischemic brain injury in the infant rat. Pediatr Res 1996; 39 (4 Pt 1): 591-597
  • 26 Takata T, Nabetani M, Okada Y. Effects of hypothermia on the neuronal activity, [Ca2+]i accumulation and ATP levels during oxygen and/or glucose deprivation in hippocampal slices of guinea pigs. Neurosci Lett 1997; 227 (01) 41-44
  • 27 McManus T, Sadgrove M, Pringle AK, Chad JE, Sundstrom LE. Intraischaemic hypothermia reduces free radical production and protects against ischaemic insults in cultured hippocampal slices. J Neurochem 2004; 91 (02) 327-336
  • 28 Rocha-Ferreira E, Vincent A, Bright S, Peebles DM, Hristova M. The duration of hypothermia affects short-term neuroprotection in a mouse model of neonatal hypoxic ischaemic injury. PLoS One 2018; 13 (07) e0199890
  • 29 Perrone S, Weiss MD, Proietti F. et al. Identification of a panel of cytokines in neonates with hypoxic ischemic encephalopathy treated with hypothermia. Cytokine 2018; 111: 119-124
  • 30 Chevin M, Guiraut C, Sébire G. Effect of hypothermia on interleukin-1 receptor antagonist pharmacodynamics in inflammatory-sensitized hypoxic-ischemic encephalopathy of term newborns. J Neuroinflammation 2018; 15 (01) 214
  • 31 Zhou T, Lin H, Jiang L. et al. Mild hypothermia protects hippocampal neurons from oxygen-glucose deprivation injury through inhibiting caspase-3 activation. Cryobiology 2018; 80: 55-61
  • 32 Dominici M, Le Blanc K, Mueller I. et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006; 8 (04) 315-317
  • 33 Ding M, Shen Y, Wang P. et al. Exosomes isolated from human umbilical cord mesenchymal stem cells alleviate neuroinflammation and reduce amyloid-beta deposition by modulating microglial activation in Alzheimer's disease. Neurochem Res 2018; 43 (11) 2165-2177
  • 34 Mukai T, Tojo A, Nagamura-Inoue T. Umbilical cord-derived mesenchymal stromal cells contribute to neuroprotection in neonatal cortical neurons damaged by oxygen-glucose deprivation. Front Neurol 2018; 9: 466
  • 35 Guo ZY, Sun X, Xu XL, Zhao Q, Peng J, Wang Y. Human umbilical cord mesenchymal stem cells promote peripheral nerve repair via paracrine mechanisms. Neural Regen Res 2015; 10 (04) 651-658
  • 36 Mukai T, Mori Y, Shimazu T. et al. Intravenous injection of umbilical cord-derived mesenchymal stromal cells attenuates reactive gliosis and hypomyelination in a neonatal intraventricular hemorrhage model. Neuroscience 2017; 355: 175-187
  • 37 Sung DK, Sung SI, Ahn SY, Chang YS, Park WS. Thrombin preconditioning boosts biogenesis of extracellular vesicles from mesenchymal stem cells and enriches their cargo contents via protease-activated receptor-mediated signaling pathways. Int J Mol Sci 2019; 20 (12) E2899
  • 38 Dalous J, Pansiot J, Pham H. et al. Use of human umbilical cord blood mononuclear cells to prevent perinatal brain injury: a preclinical study. Stem Cells Dev 2013; 22 (01) 169-179
  • 39 Archambault J, Moreira A, McDaniel D, Winter L, Sun L, Hornsby P. Therapeutic potential of mesenchymal stromal cells for hypoxic ischemic encephalopathy: a systematic review and meta-analysis of preclinical studies. PLoS One 2017; 12 (12) e0189895
  • 40 Thoresen M. Combining two good treatments makes it worse. Brain Behav Immun 2018; 71: 7-8
  • 41 Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 2005; 308 (5726): 1314-1318
  • 42 Li J, Yawno T, Sutherland A. et al. Preterm white matter brain injury is prevented by early administration of umbilical cord blood cells. Exp Neurol 2016; 283 (pt. A): 179-187
  • 43 Kim S, Kim YE, Hong S. et al. Reactive microglia and astrocytes in neonatal intraventricular hemorrhage model are blocked by mesenchymal stem cells. Glia 2020; 68 (01) 178-192
  • 44 Mukai T, Martino ED, Tsuji S. et al. Umbilical cord tissue-derived mesenchymal stromal cells immunomodulate and restore actin dynamics and phagocytosis of lipopolysaccharide-activated microglia via the PI3K/Akt/Rho GTPase pathway, with lot-to-lot variation. Cell Death Disc 2021; 7: 46
  • 45 Nagamura-Inoue T, Mukai T. Umbilical cord is a rich source of mesenchymal stromal cells for cell therapy. Curr Stem Cell Res Ther 2016; 11 (08) 634-642
  • 46 Krampera M, Glennie S, Dyson J. et al. Bone marrow mesenchymal stem cells inhibit the response of naive and memory antigen-specific T cells to their cognate peptide. Blood 2003; 101 (09) 3722-3729
  • 47 Weiss ML, Anderson C, Medicetty S. et al. Immune properties of human umbilical cord Wharton's jelly-derived cells. Stem Cells 2008; 26 (11) 2865-2874
  • 48 Aridas JD, McDonald CA, Paton MC. et al. Cord blood mononuclear cells prevent neuronal apoptosis in response to perinatal asphyxia in the newborn lamb. J Physiol 2016; 594 (05) 1421-1435
  • 49 Hattori T, Sato Y, Kondo T. et al. Administration of umbilical cord blood cells transiently decreased hypoxic-ischemic brain injury in neonatal rats. Dev Neurosci 2015; 37 (02) 95-104
  • 50 Yoshihara T, Taguchi A, Matsuyama T. et al. Increase in circulating CD34-positive cells in patients with angiographic evidence of moyamoya-like vessels. J Cereb Blood Flow Metab 2008; 28 (06) 1086-1089
  • 51 Rosenkranz K, Kumbruch S, Lebermann K. et al. The chemokine SDF-1/CXCL12 contributes to the ‘homing’ of umbilical cord blood cells to a hypoxic-ischemic lesion in the rat brain. J Neurosci Res 2010; 88 (06) 1223-1233
  • 52 Yasuhara T, Hara K, Maki M. et al. Mannitol facilitates neurotrophic factor up-regulation and behavioural recovery in neonatal hypoxic-ischaemic rats with human umbilical cord blood grafts. J Cell Mol Med 2010; 14 (04) 914-921
  • 53 Majka M, Janowska-Wieczorek A, Ratajczak J. et al. Numerous growth factors, cytokines, and chemokines are secreted by human CD34(+) cells, myeloblasts, erythroblasts, and megakaryoblasts and regulate normal hematopoiesis in an autocrine/paracrine manner. Blood 2001; 97 (10) 3075-3085
  • 54 Taguchi A, Matsuyama T, Moriwaki H. et al. Circulating CD34-positive cells provide an index of cerebrovascular function. Circulation 2004; 109 (24) 2972-2975
  • 55 Taguchi A, Soma T, Tanaka H. et al. Administration of CD34+ cells after stroke enhances neurogenesis via angiogenesis in a mouse model. J Clin Invest 2004; 114 (03) 330-338
  • 56 Nakatomi H, Kuriu T, Okabe S. et al. Regeneration of hippocampal pyramidal neurons after ischemic brain injury by recruitment of endogenous neural progenitors. Cell 2002; 110 (04) 429-441
  • 57 Drago J, Murphy M, Carroll SM, Harvey RP, Bartlett PF. Fibroblast growth factor-mediated proliferation of central nervous system precursors depends on endogenous production of insulin-like growth factor I. Proc Natl Acad Sci U S A 1991; 88 (06) 2199-2203
  • 58 Kasahara Y, Yamahara K, Soma T. et al. Transplantation of hematopoietic stem cells: intra-arterial versus intravenous administration impacts stroke outcomes in a murine model. Transl Res 2016; 176: 69-80
  • 59 Kikuchi-Taura A, Okinaka Y, Takeuchi Y. et al. Bone marrow mononuclear cells activate angiogenesis via GAP junction-mediated cell-cell interaction. Stroke 2020; 51 (04) 1279-1289
  • 60 Felling RJ, Snyder MJ, Romanko MJ. et al. Neural stem/progenitor cells participate in the regenerative response to perinatal hypoxia/ischemia. J Neurosci 2006; 26 (16) 4359-4369
  • 61 Eglitis MA, Mezey E. Hematopoietic cells differentiate into both microglia and macroglia in the brains of adult mice. Proc Natl Acad Sci U S A 1997; 94 (08) 4080-4085
  • 62 Mezey E, Key S, Vogelsang G, Szalayova I, Lange GD, Crain B. Transplanted bone marrow generates new neurons in human brains. Proc Natl Acad Sci U S A 2003; 100 (03) 1364-1369
  • 63 Li Y, Adomat H, Guns ET. et al. Identification of a hematopoietic cell dedifferentiation-inducing factor. J Cell Physiol 2016; 231 (06) 1350-1363
  • 64 Chen SH, Wang JJ, Chen CH. et al. Umbilical cord blood-derived CD34+ cells improve outcomes of traumatic brain injury in rats by stimulating angiogenesis and neurogenesis. Cell Transplant 2014; 23 (08) 959-979
  • 65 Davoust N, Vuaillat C, Cavillon G. et al. Bone marrow CD34+/B220+ progenitors target the inflamed brain and display in vitro differentiation potential toward microglia. FASEB J 2006; 20 (12) 2081-2092
  • 66 Perlman JM, Wyllie J, Kattwinkel J. et al; Neonatal Resuscitation Chapter Collaborators. Part 11: Neonatal resuscitation: 2010 International Consensus on Cardiopulmonary Resuscitation and Emergency Cardiovascular Care Science With Treatment Recommendations. Circulation 2010; 122 (16, Suppl 2): S516-S538
  • 67 Cotten CM, Murtha AP, Goldberg RN. et al. Feasibility of autologous cord blood cells for infants with hypoxic-ischemic encephalopathy. J Pediatr 2014; 164 (05) 973-979.e1
  • 68 Tsuji M, Sawada M, Watabe S. et al. Autologous cord blood cell therapy for neonatal hypoxic-ischaemic encephalopathy: a pilot study for feasibility and safety. Sci Rep 2020; 10 (01) 4603
  • 69 Mukai T, Tojo A, Nagamura-Inoue T. Mesenchymal stromal cells as a potential therapeutic for neurological disorders. Regen Ther 2018; 9: 32-37
  • 70 Huang L, Zhang C, Gu J. et al. A randomized, placebo-controlled trial of human umbilical cord blood mesenchymal stem cell infusion for children with cerebral palsy. Cell Transplant 2018; 27 (02) 325-334
  • 71 Wang X, Hu H, Hua R. et al. Effect of umbilical cord mesenchymal stromal cells on motor functions of identical twins with cerebral palsy: pilot study on the correlation of efficacy and hereditary factors. Cytotherapy 2015; 17 (02) 224-231
  • 72 Dong H, Li G, Shang C. et al. Umbilical cord mesenchymal stem cell (UC-MSC) transplantations for cerebral palsy. Am J Transl Res 2018; 10 (03) 901-906
  • 73 Boruczkowski D, Zdolińska-Malinowska I. Wharton's Jelly mesenchymal stem cell administration improves quality of life and self-sufficiency in children with cerebral palsy: results from a retrospective study. Stem Cells Int 2019; 2019: 7402151
  • 74 Fu X, Hua R, Wang X. et al. Synergistic improvement in children with cerebral palsy who underwent double-course human Wharton's Jelly stem cell transplantation. Stem Cells Int 2019; 2019: 7481069-7481069
  • 75 Gu J, Huang L, Zhang C. et al. Therapeutic evidence of umbilical cord-derived mesenchymal stem cell transplantation for cerebral palsy: a randomized, controlled trial. Stem Cell Res Ther 2020; 11 (01) 43
  • 76 Cheng H, Liu X, Hua R. et al. Clinical observation of umbilical cord mesenchymal stem cell transplantation in treatment for sequelae of thoracolumbar spinal cord injury. J Transl Med 2014; 12: 253
  • 77 Jin JL, Liu Z, Lu ZJ. et al. Safety and efficacy of umbilical cord mesenchymal stem cell therapy in hereditary spinocerebellar ataxia. Curr Neurovasc Res 2013; 10 (01) 11-20
  • 78 Wang S, Cheng H, Dai G. et al. Umbilical cord mesenchymal stem cell transplantation significantly improves neurological function in patients with sequelae of traumatic brain injury. Brain Res 2013; 1532: 76-84
  • 79 Li JF, Zhang DJ, Geng T. et al. The potential of human umbilical cord-derived mesenchymal stem cells as a novel cellular therapy for multiple sclerosis. Cell Transplant 2014; 23 (Suppl. 01) S113-S122
  • 80 Riordan NH, Morales I, Fernández G. et al. Clinical feasibility of umbilical cord tissue-derived mesenchymal stem cells in the treatment of multiple sclerosis. J Transl Med 2018; 16 (01) 57
  • 81 Lv YT, Zhang Y, Liu M. et al. Transplantation of human cord blood mononuclear cells and umbilical cord-derived mesenchymal stem cells in autism. J Transl Med 2013; 11: 196
  • 82 Riordan NH, Hincapié ML, Morales I. et al. Allogeneic human umbilical cord mesenchymal stem cells for the treatment of autism spectrum disorder in children: safety profile and effect on cytokine levels. Stem Cells Transl Med 2019; 8 (10) 1008-1016
  • 83 Bélanger M, Allaman I, Magistretti PJ. Brain energy metabolism: focus on astrocyte-neuron metabolic cooperation. Cell Metab 2011; 14 (06) 724-738